You are on page 1of 30

Biotechnology Advances 24 (2006) 452 481 www.elsevier.

com/locate/biotechadv

Research review paper

Outlook for cellulase improvement: Screening and selection strategies


Y.-H. Percival Zhang a,, Michael E. Himmel b , Jonathan R. Mielenz c
a

Biological Systems Engineering Department, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA b National Bioenergy Center, National Renewable Energy Laboratory, Golden, CO 80401, USA c Life Science Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA Received 31 January 2006; received in revised form 6 March 2006; accepted 11 March 2006 Available online 27 March 2006

Abstract Cellulose is the most abundant renewable natural biological resource, and the production of biobased products and bioenergy from less costly renewable lignocellulosic materials is important for the sustainable development of human beings. A reduction in cellulase production cost, an improvement in cellulase performance, and an increase in sugar yields are all vital to reduce the processing costs of biorefineries. Improvements in specific cellulase activities for non-complexed cellulase mixtures can be implemented through cellulase engineering based on rational design or directed evolution for each cellulase component enzyme, as well as on the reconstitution of cellulase components. Here, we review quantitative cellulase activity assays using soluble and insoluble substrates, and focus on their advantages and limitations. Because there are no clear relationships between cellulase activities on soluble substrates and those on insoluble substrates, soluble substrates should not be used to screen or select improved cellulases for processing relevant solid substrates, such as plant cell walls. Cellulase improvement strategies based on directed evolution using screening on soluble substrates have been only moderately successful, and have primarily targeted improvement in thermal tolerance. Heterogeneity of insoluble cellulose, unclear dynamic interactions between insoluble substrate and cellulase components, and the complex competitive and/or synergic relationship among cellulase components limit rational design and/or strategies, depending on activity screening approaches. Herein, we hypothesize that continuous culture using insoluble cellulosic substrates could be a powerful selection tool for enriching beneficial cellulase mutants from the large library displayed on the cell surface. 2006 Elsevier Inc. All rights reserved.
Keywords: Cellulase activity assay; Cellulose; Cellulosome; Continuous culture; Enzymatic cellulose hydrolysis; High throughput screening; Selection; Sugar assay

Abbreviations: AFEX, ammonia fiber explosion; BC, bacterial cellulose; BCA, 2,2-bicinchroninate; BMCC, bacterial microcrystalline cellulose; CMC, carboxymethyl cellulose; CBM, cellulose-binding module; CBP, consolidated bioprocessing; CrI, crystallinity index; DMAc, N,Ndimethylacetamide; DNS, dinitrosalicyclic acid; DP, degree of polymerization of cellulose; DS, degree of substitution; DTT, dithiothreitol; Fa, fraction of -glucosidic bond accessible to cellulase; FPA, filter paper activity; FRE, fraction of the reducing end to all anhydroglucose units of cellulose, 1/DP; HEC, hydroxyethyl cellulose; PASC, phosphoric acid swollen cellulose; RAC, regenerated amorphous cellulose; PAHBAH, 4hydroxybenzoylhydrazine; RS, selection ratio; TNP-CMC, trinitrophenyl-carboxymethyl cellulose. Corresponding author. Tel.: +1 540 231 7414; fax: +1 540 231 3199. E-mail address: ypzhang@vt.edu (Y.-H. Percival Zhang). 0734-9750/$ - see front matter 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.biotechadv.2006.03.003

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

453

Contents Introduction . . . . . . . . . . . . . . . . . . Cellulose hydrolysis mechanisms . . . . . . . Substrates for cellulase activity assays . . . . . 3.1. Soluble substrates . . . . . . . . . . . . 3.2. Insoluble substrates . . . . . . . . . . . 4. Quantitative assays . . . . . . . . . . . . . . . 4.1. Hydrolysis products . . . . . . . . . . . 4.2. Cellulase activity assays. . . . . . . . . 4.2.1. Endoglucanases . . . . . . . . 4.2.2. Exoglucanases . . . . . . . . . 4.2.3. -D-glucosidases . . . . . . . . 4.2.4. Total cellulase . . . . . . . . . 5. Cellulase improvement and screening/selection 5.1. Rational design . . . . . . . . . . . . . 5.2. Directed evolution. . . . . . . . . . . . 5.3. Screening . . . . . . . . . . . . . . . . 5.4. Selection . . . . . . . . . . . . . . . . 6. Summary . . . . . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . 1. 2. 3. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453 455 457 458 458 460 461 462 462 463 464 464 465 465 467 469 470 472 472 472

1. Introduction Cellulose is the primary product of photosynthesis in terrestrial environments, and the most abundant renewable bioresource produced in the biosphere ( 100 billion dry tons/year) (Holtzapple, 1993; Jarvis, 2003; Zhang and Lynd, 2004b). Cellulose biodegradation by cellulases and cellulosomes, produced by numerous microorganisms, represents a major carbon flow from fixed carbon sinks to atmospheric CO2 (Berner, 2003; Falkowski et al., 2000; Melillo et al., 2002), is very important in several agricultural and waste treatment processes (Angenent et al., 2004; Das and Singh, 2004; Haight, 2005; Hamer, 2003; Humphrey et al., 1977; Russell and Rychlik, 2001; Schloss et al., 2005; van Wyk, 2001), and could be widely used to produce sustainable biobased products and bioenergy to replace depleting fossil fuels (Angenent et al., 2004; Demain et al., 2005; Galbe and Zacchi, 2002; Hall et al., 1993; Hoffert et al., 2002; Kamm and Kamm, 2004; Lynd, 1996; Lynd et al., 1991, 2002, 1999; Mielenz, 2001; Mohanty et al., 2000; Moreira, 2005; Reddy and Yang, 2005; Wyman, 1994, 1999, 2003). Additionally, studies have shown that the use of biobased products and bioenergy can achieve zero net carbon dioxide emission (Demain, 2004; Demain et al., 2005; Hoffert et al., 2002; Lynd et al., 1991, 1999). Development of technologies for effectively converting less costly agricultural and forestry residues to fermentable sugars

offers outstanding potential to benefit the national interest through: (1) improved strategic security, (2) decreased trade deficits, (3) healthier rural economies, (4) improved environmental quality, (5) technology exports, and (6) a sustainable energy resource supply (Angenent et al., 2004; Caldeira et al., 2003; Demain et al., 2005; Hoffert et al., 1998, 2002; Kamm and Kamm, 2004; Lynd, 1996; Lynd et al., 1991, 1999, 2002; Moreira, 2005; Wirth et al., 2003; Wyman, 1999). Effective conversion of recalcitrant lignocellulose to fermentable sugars requires three sequential steps: (1) size reduction, (2) pretreatment/fractionation, and (3) enzymatic hydrolysis (Wyman, 1999; Zhang and Lynd, 2004b). One of the most important and difficult technological challenges is to overcome the recalcitrance of natural lignocellulosic materials, which must be enzymatically hydrolyzed to produce fermentable sugars (Chang et al., 1981; Demain et al., 2005; Fan et al., 1982; Grethlein, 1984; Hsu, 1996; Lin et al., 1981; McMillian, 1994; Millett et al., 1976; Moreira, 2005; Mosier et al., 2005; Saddler et al., 1993; Weil et al., 1994; Wyman, 1999; Wyman et al., 2005a). Cellulases are relatively costly enzymes, and a significant reduction in cost will be important for their commercial use in biorefineries. Cellulase-based strategies that will make the biorefinery processing more economical include: increasing commercial enzyme volumetric productivity, producing enzymes using

454

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

cheaper substrates, producing enzyme preparations with greater stability for specific processes, and producing cellulases with higher specific activity on solid substrates. Recently, the biotechnology companies Genencor International and Novozymes Biotech have reported the development of technology that has reduced the cellulase cost for the cellulose-to-ethanol process from US$5.40 per gallon of ethanol to approximately 20 cents per gallon of ethanol (Moreira, 2005), in which the two main strategies were (1) an economical improvement in production of cellulase to reduce US$ per gram of enzyme by process and strain enhancement, e.g., cheaper medium from lactose to glucose and alternative inducer system and (2) an improvement in the cellulase enzyme performance to reduce grams of enzyme for achieving equivalent hydrolysis by cocktails and component improvement (Knauf and Moniruzzaman, 2004). But this claim has not yet been widely accepted because the cellulase mixture was tested only for the specific pretreated lignocellulosic substrate and cannot be applied to other pretreated lignocelluloses. Currently, most commercial cellulases (including glucosidase) are produced by Trichoderma species and Aspergillus species (Cherry and Fidantsef, 2003; Esterbauer et al., 1991; Kirk et al., 2002). Cellulases are used in the textile industry for cotton softening and denim finishing; in the detergent market for color care, cleaning, and anti-deposition; in the food industry for mashing; and in the pulp and paper industries for de-

inking, drainage improvement, and fiber modification (Cherry and Fidantsef, 2003; Kirk et al., 2002). The cellulase market is expected to expand dramatically when cellulases are used to hydrolyze pretreated cellulosic materials to sugars, which can be fermented to commodities such as bioethanol and biobased products on a large scale (Cherry and Fidantsef, 2003; Himmel et al., 1999; van Beilen and Li, 2002). For example, the potential cellulase market has been estimated to be as high as US$400 million per year if cellulases are used for hydrolyzing the available corn stover in the midwestern United States (van Beilen and Li, 2002). This market scenario represents an increase of 33% in the total US industrial enzyme market (Wolfson, 2005). The large market potential and the important role that cellulases play in the emerging bioenergy and bio-based products industries provide a great motivation to develop better cellulase preparations for plant cell wall cellulose hydrolysis. These improved cellulases must also have characteristics necessary for biorefineries, such as higher catalytic efficiency on insoluble cellulosic substrates, increased stability at elevated temperature and at a certain pH, and higher tolerance to end-product inhibition. Fig. 1 shows that cellulase engineering for noncomplexed cellulase systems contains three major research directions: (1) rational design for each cellulase, based on knowledge of the cellulase structure and the catalytic mechanism (Schulein, 2000; Wilson, 2004; Wither, 2001); (2) directed evolution for each

Wild type Cellulase Components Reconstitute cellulase cocktail endos exosR expsNR -Gase Improved cellulase components

Rational Design

Directed Evolution

Screen or select on solid substrate

Fig. 1. Scheme of cellulase engineering for non-complexed cellulases. Endos, endoglucanases; exosR, exoglucanases acting on reducing ends; exosNR, exoglucanases acting on non-reducing ends; -Gase, -glucosidase.

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

455

cellulase, in which the improved enzymes or ones with new properties were selected or screened after random mutagenesis and/or molecular recombination (Arnold, 2001; Cherry and Fidantsef, 2003; Hibbert et al., 2005; Schmidt-Dannert and Arnold, 1999; Shoemaker et al., 2003; Tao and Cornish, 2002); and (3) the reconstitution of cellulase mixtures (cocktails) active on insoluble cellulosic substrates, yielding an improved hydrolysis rate or higher cellulose digestibility (Baker et al., 1998; Boisset et al., 2001; Himmel et al., 1999; Irwin et al., 1993; Kim et al., 1998; Sheehan and Himmel, 1999; Walker et al., 1993; Wilson and Walker, 1991; Zhang and Lynd, 2004b). With respect to engineering complexed cellulase systems (cellulosomes), the idea of chimeric constructs of cellulosomal domains/components was proposed by Bayer et al. (1994), and the reconstruction of cellulosome components is becoming another hot research area (Fierobe et al., 2001, 2002, 2005; Mingardon et al., 2005; Sabathe and Soucaille, 2003), which we do not review here. The cornerstone of enzyme engineering is to achieve a direct correlation between the enzyme assays or screening approaches and the changes in enzyme functions in the desired application. Development of a useful, predictive cellulase assay or screening is particularly difficult because of the nature of solid heterogeneous substrates, such as plant cell walls. Available quantitative cellulase assays and screenings have been analyzed and compared herein, including their advantages and limitations. Also, successful cellulase examples using directed evolution are examined, and a possible strategy of combinatorial molecular breeding and continuous culture with solid cellulosic materials to select a cellulase with higher activity is discussed. 2. Cellulose hydrolysis mechanisms Cellulose is a linear condensation polymer consisting of D-anhydroglucopyranose joined together by -1,4glycosidic bonds with a degree of polymerization (DP) from 100 to 20,000 (Krassig, 1993; O'Sullivan, 1997; Tomme et al., 1995; Zhang and Lynd, 2004b). Anhydrocellobiose is the repeating unit of cellulose. Coupling of adjacent cellulose chains and sheets of cellulose by hydrogen bonds and van der Waal's forces results in a parallel alignment and a crystalline structure with straight, stable supra-molecular fibers of great tensile strength and low accessibility (Demain et al., 2005; Krassig, 1993; Nishiyama et al., 2003; Notley et al., 2004; Zhang and Lynd, 2004b; Zhbankov, 1992). The cellulose molecule is very stable, with a half life of

58 million years for -glucosidic bond cleavage at 25 C (Wolfenden and Snider, 2001), while the much faster enzyme-driven cellulose biodegradation process is vital to return the carbon in sediments to the atmosphere (Berner, 2003; Cox et al., 2000; Falkowski et al., 2000; Schlamadinger and Marland, 1996). The widely accepted mechanism for enzymatic cellulose hydrolysis involves synergistic actions by endoglucanase (EC 3.2.1.4), exoglucanase or cellobiohydrolase (EC 3.2.1.91), and -glucosidase (EC 3.2.1.21) (Henrissat, 1994; Knowles et al., 1987; Lynd et al., 2002; Teeri, 1997; Wood and GaricaCampayo, 1990; Zhang and Lynd, 2004b). Endoglucanases hydrolyze accessible intramolecular -1,4glucosidic bonds of cellulose chains randomly to produce new chain ends; exoglucanases processively cleave cellulose chains at the ends to release soluble cellobiose or glucose; and -glucosidases hydrolyze cellobiose to glucose in order to eliminate cellobiose inhibition. These three hydrolysis processes occur simultaneously as shown in Fig. 2. Primary hydrolysis that occurs on the surface of solid substrates releases soluble sugars with a degree of polymerization (DP) up to 6 into the liquid phase upon hydrolysis by endoglucanases and exoglucanases. The enzymatic depolymerization step performed by endoglucanases and exoglucanases is the rate-limiting step for the whole cellulose hydrolysis process. Secondary hydrolysis that occurs in the liquid phase involves primarily the hydrolysis of cellobiose to glucose by -glucosidases, although some -glucosidases also hydrolyze longer cellodextrins (Zhang and Lynd, 2004b). During cellulose hydrolysis, the solid substrate characteristics vary, including (1) changes in the cellulose chain end number resulting from generation by endoglucanases and consumption by exoglucanases (Kleman-Leyer et al., 1992, 1994, 1996; Kongruang et al., 2004; Srisodsuk et al., 1998; Zhang and Lynd, 2005b) and (2) changes in cellulose accessibility resulting from substrate consumption and cellulose fragmentation (Banka et al., 1998; Boisset et al., 2000; Chanzy et al., 1983; Din et al., 1991, 1994; Halliwell and Riaz, 1970; Lee et al., 1996, 2000; Saloheimo et al., 2002; Walker et al., 1990, 1992; Wang et al., 2003; Woodward et al., 1992). The combined actions of endoglucanases and exoglucanases modify the cellulose surface characteristics (topography) over time, resulting in rapid changes in hydrolysis rates. The complicated interactions among endoglucanases, exoglucanases, and the changing substrate characteristics during hydrolysis have been simulated by a new functionally based mathematical model

456

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

Solid Phase

(primary hydrolysis)

Liquid Phase (secondary hydrolysis)

endos exosR exosNR -Gase

Fig. 2. Mechanistic scheme of enzymatic cellulose hydrolysis by Trichoderma non-complexed cellulase system.

(Zhang and Lynd, in press), applying a set of enzymatic parameters for endoglucanase I, cellobiohydrolases I and II to a variety of substrates with two important substrate properties: the fraction of -glucosidic bond accessible to cellulase (Fa) (Zhang and Lynd, 2004b) and the degree of polymerization (DP) (Okazaki and Moo-Young, 1978; Zhang and Lynd, 2004b) (see Table 1). In this way, disparate information from the

literature was framed in a coherent way to facilitate an understanding of enzymatic cellulose hydrolysis. For example, the reaction rates simulated by the model were consistent with a substantial number of observations reported in the literature, including the effects of substrate characteristics on exoglucanase and endoglucanase activities; the effects of substrate characteristics and experimental conditions on the degree of

Table 1 Summary of typical values of model celluloses for crystallinity index (CrI), the fraction of -glucosidic bond accessible to cellulase (Fa), which is estimated by maximum cellulase adsorption capacity (Zhang and Lynd, 2004b), the number average of degree of polymerization (DPN), the fraction of reducing ends (FRE), and relative ratio of FRE/Fa Substrates CrI Fa (%) DPN FRE (%) Low 26 1002000 10003000 7502800 6002000 150500 1001000 7501500 4001000 16.67 0.05 High 50 1 FRE/Fa Low 0.167 0.0005 High 0.5 0.01

Soluble Cellodextrins and their derivatives CMC Insoluble Cotton Whatman No. 1 filter paper Bacterial cellulose Microcrytalline cellulose (Avicel) PASC Pulp (Solka Floc) Pretreated cellulosic substrates

N.A. N.A.

100 100

0.80.95 0.45 0.80.95 0.50.6 0 0.40.7 0.40.7

0.2 1.8 6 0.6 12 1.8 0.6

0.033 0.036 0.05 0.2 0.1 0.067 0.1

0.1 0.133 0.167 0.667 1 0.133 0.25

0.167 0.0198 0.00833 0.333 0.00833 0.0370 0.167

0.5 0.0741 0.0278 1.11 0.0833 0.0741 0.417

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

457

endo-exo synergy; the effects of endoglucanase partition coefficient on the hydrolysis rates; and the effects of enzyme loading on relative reaction rates for different substrates. The model also suggests that it is nearly impossible to predict hydrolysis performance of cellulase mixtures from one solid substrate to another solid substrate, because of large variations in total cellulase concentration, ratio of endo/exocellulases, reaction time, and substrate characteristics. Therefore, enzyme reconstitution may have be conducted so as to achieve better performance for a specific substrate (Knauf and Moniruzzaman, 2004). Unlike non-complexed fungal cellulase, anaerobic microorganisms possess complexed cellulase systems, called cellulosomes (Bayer et al., 1994, 1998, 2004; Beguin and Alzari, 1998; Demain et al., 2005; Doi and Kosugi, 2004; Doi et al., 1998; Doi and Tamaru, 2001; Leschine, 1995; Schwarz, 2001). Leschine (1995) estimated that anaerobic cellulose degradation could account for only 510% of total cellulose biodegradation, but it could be underestimated because anaerobic cellulose hydrolysis is responsible for considerable carbon recycling in the anoxic zones of ponds, lakes, oceans, and intestines of ruminants and guts of termites (P.J. Weimer, personal communication). Furthermore, an understanding anaerobic cellulase systems are of significant importance to basic

sciences, such as the evolution of cellulase genes, the structures of cellulases, and the formation and hydrolysis of reacting biofilms on cellulose surfaces (Lynd et al., in press). Anaerobic cellulose fermentation has both current and future applications, such as agricultural processes anaerobic waste treatment, and consolidated bioprocessing (CBP), respectively (Lynd, 1996; Lynd et al., 1999, 2002, 2005). Recently, a microbial cellulose hydrolysis mechanism has been reported for the anaerobic cellulolytic bacterium Clostridium thermocellum that assimilates longer soluble hydrolysis products with an average degree of polymerization of 4 rather than glucose and cellobiose. The improved bioenergetics resulting from longer chain sugar assimilation supports the biological feasibility of anaerobic fermentation without added saccharolytic enzymes (Zhang and Lynd, 2005c). More information about the cellulosomebased microbial cellulose hydrolysis research is available elsewhere (Lynd, 1996; Lynd et al., 2002, 1999, 2005; Zhang and Lynd, 2003a, 2004a, 2005a). 3. Substrates for cellulase activity assays Substrates for cellulase activity assays can be divided into two categories, based on their solubility in water (Table 2).

Table 2 Substrates containing -1,4-glucosidic bonds hydrolyzed by cellulases and their detections Substrate Soluble Short chain (low DP) Cellodextrins Radio-labeled cellodextrins Cellodextrin derivatives -methylumbelliferyl-oligosaccharides p-nitrophenol-oligosaccharides Long chain cellulose derivatives Carboxymethyl cellulose (CMC), hydroxyethyl cellulose (HEC) Dyed CMC Insoluble Crystalline celluloseCotton, microcrystalline cellulose (Avicel), Valonia cellulose, bacterial cellulose Amorphous cellulose - PASC, alkali-swollen cellulose RAC Dyed cellulose Fluorescent cellulose Chromogenic and fluorephoric derivatives Trinitrophenyl-carboxymethylcellulose (TNP-CMC) Fluram-cellulose Practical cellulose-containing substrates -cellulose, pretreated lignocellulosic biomass
a

Detection a

Enzymes

RS, HPLC; TLC TLC plus liquid scintillation Fluorophore liberation, TLC Chromophore liberation, TLC RS; viscosity Dye liberation

Endo, Exo, BG Endo, Exo, BG Endo, Exo, BG Endo, Exo, BG Endo Endo

RS, TSS, HPLC RS, TSS, HPLC RS, TSS, HPLC, TLC Dye liberation Fluorophore liberation Chromophore liberation Fluorophore liberation HPLC, RS

Total, Endo, Exo Total, Endo, Exo Total, Endo Total Endo Endo, Total Total

RS, reducing sugars; TSS, total soluble sugars.

458

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

3.1. Soluble substrates Soluble substrates include low DP cellodextrins from 2 to 6 sugar units and their derivatives, as well as long DP cellulose derivatives (ca. several hundreds of sugar units). They are often used for measuring individual cellulase component activity (Table 2). Cellodextrins are soluble for DP 6, and very slightly soluble for 6 < DP < 12 (Miller, 1960; Miller, 1963; Pereira et al., 1988; Zhang and Lynd, 2003b, 2005b). Their solubility decreases drastically with increasing DP because of strong intermolecular hydrogen bonds and system entropic effects. Cellodextrins are often prepared through cellulose hydrolysis by fuming HCl (Miller, 1960, 1963), sulfuric acid (Voloch et al., 1984), acetylation (Schmid et al., 1988; Wolfram and Dacons, 1952), or mixed acids (HCl and H2SO4) (Zhang and Lynd, 2003b). Cellodextrins are also prepared through biosynthesis using C. thermocellum cellobiose and cellodextrin phosphorylases (Ng and Zeikus, 1986; Strobel et al., 1995; Zhang and Lynd, 2005c, 2006), or T. reesei -glucosidase (Chirico and Brown, 1987). Cellodextrin mixtures can be separated into single components using chromatographic methods such as charcoal-celite (Miller, 1963), thin layer (Chirico and Brown, 1985; Zhang and Lynd, 2006), cation exchange (Huebner et al., 1978; Voloch et al., 1984), or sizeexclusion (Schmid et al., 1988; Shintate et al., 2003; Zhang and Lynd, 2003b, 2006). Chromogenic p-nitrophenyl glycosides and fluorogenic methylumbelliferyl-D-glycosides derived from soluble cellodextrins are very useful for the study of initial cellulase kinetics (Tuohy et al., 2002; Wolfgang and Wilson, 1999), reaction specificity (Bhat et al., 1990; Claeyssens and Aerts, 1992; Tomme et al., 1996; van Tilbeurgh and Claeyssens, 1985; van Tilbeurgh et al., 1982, 1985; Zverlov et al., 2002b), and binding site thermodynamics (Barr and Holewinski, 2002). They are also used to determine the inhibition constants of cellulase in the presence of added cellobiose and glucose (Tuohy et al., 2002), because chromophores released from substituted glycosides can be easily measured independently of sugars. Long DP cellulose derivatives can be dissolved in water because of their chemical substitutions. Ionicsubstituted carboxymethyl cellulose (CMC) is often used for determining endoglucanase activity, called CMCase, because endoglucanases cleave intramolecular -1,4-glucosidic bonds randomly, resulting in a dramatic reduction in the DP (i.e., specific viscosity) of CMC. CMC has two very important physical parameters the degree of substitution (DS) and DP. The solubility

of CMC is closely associated with the DS that has a maximum stoichiometric value of 3. CMC is soluble in water when DS > 0.30.7 (Karlsson et al., 2001; Klemm et al., 1998a; Wood and Bhat, 1988). Commercial CMCs usually have a DS < 1.5. It is strongly recommended that a reducing sugar assay or viscosity assay using CMC as a substrate should be limited to the first 2% hydrolysis of substrate when DS = 0.7 (Wood and Bhat, 1988). This is important because only nonsubstituted glucose units are accessible to cellulase, and hydrolysis action requires at least two or three contiguous non-substituted residues. The DP of CMC is not important for the reducing sugar assay, but it is very important for determining viscosity reduction. CMC dissolution in water should be done by gentle swirling to avoid DP reduction (Sharrock, 1988). Also, the viscosity of ionic CMC is influenced by pH, ionic strength, and polyvalent cation concentration. Therefore, it is recommended to use nonionic substituted celluloses, such as hydroxyethyl cellulose (HEC), for determining endoglucanase activity (Wood and Bhat, 1988). Dyed soluble CMC is made by mixing CMC with dyesRemazol Brilliant Blue R (Flp and Ponyi, 1997; McCleary, 1980; Wirth and Wolf, 1992) or Ruthenium Red (Rescigno et al., 1994). The colors released from soluble cellulose derivatives in the supernatant can be measured after precipitation of the soluble substrates. A dyed CMC is also used as a substrate on solid agar plates, sometimes called zymograms (Bera-Maillet et al., 2000; Eriksson and Petterson, 1973; Holt and Hartman, 1994). After hydrolysis, staining, and washing, halo zones can be observed in the colored background. 3.2. Insoluble substrates Insoluble cellulose-containing substrates for cellulase activity assays include nearly pure celluloses (cotton linter, Whatman No. 1 filter paper, bacterial cellulose, microcrystalline cellulose, and amorphous cellulose) and impure cellulose-containing substrates (dyed cellulose, -cellulose, and pretreated lignocellulose). Native cellulose, referred to as cellulose I, has two distinct crystallite forms, I which is dominant in bacterial and algal cellulose, and I, which is dominant in higher plants (Atalla and Vanderhart, 1984). Native cellulose (cellulose I) can be converted to other crystalline forms (IIIV) by various treatments (Klein and Snodgrass, 1993; Krassig, 1993; O'Sullivan, 1997). Table 1 shows several key physical valuescrystallinity index, degree of polymerization, and cellulose

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

459

accessibility to cellulase that can be estimated based on maximum cellulase adsorption (Zhang and Lynd, 2004b). The crystallinity index (CrI) of cellulose, quantitatively measured from its wide range X-ray diffraction pattern (Krassig, 1993; Ramos et al., 2005; Zhang and Lynd, 2004b), is not strongly associated with hydrolysis rates (Converse, 1993; Mansfield et al., 1999; Zhang and Lynd, 2004b). Nevertheless, it is still a convenient indicator representing the change in cellulose characteristics for one material before and after treatment. Cotton, bacterial cellulose, and the Valonia ventricosa algal cellulose are examples of highly crystalline cellulose (Boisset et al., 1999; Fierobe et al., 2002), whereas amorphous cellulose is at the other extreme. Microcrystalline cellulose, filter paper, -cellulose, and pretreated cellulosic substrates have modest CrI values, and can be regarded as a combination of crystalline fraction and amorphous fraction, but there is no clear borderline between two fractions. Cotton fiber is made from natural cotton after impurities, such as wax, pectin, and colored matter, have been removed (Wood, 1988). Whatman No.1 filter paper is made from long fiber cotton pulp with a low CrI = 45% (Dong et al., 1998; Henrissat et al., 1985). Microcrystalline cellulose, called hydrocellulose or avicel (the commercial name), can be purchased from several companies, such as FMC, Merck, and Sigma. It is made through the following steps: hydrolysis of wood pulp by dilute hydrochloric acid to remove the amorphous cellulose fraction, formation of colloidal dispersions by high shear fields, followed by spray drying of the washed pulp slurry (Fleming et al., 2001; Zhang and Lynd, 2004b). However, microcrystalline cellulose still contains a significant fraction of amorphous cellulose. Avicel is a good substrate for exoglucanase activity assay, because it has a low DP and relatively low accessibility (i.e., the highest ratio of FRE/Fa) (Table 1). Therefore, some researchers feel that avicelase activity is equivalent to exoglucanase activity (Wood and Bhat, 1988). However, some endoglucanases can release considerable reducing sugars from avicel (Zhang and Lynd, 2004b). Bacterial cellulose (BC) is prepared from the pellicle produced by Acetobacter xylinum (ATCC 23769) (Hestrin, 1963) or from Nata de Coco (Daiwa Fine Produces, Singapore) (Boisset et al., 2000). Bacterial microcrystalline cellulose (BMCC) can be prepared from BC by partial acid hydrolysis to remove the amorphous cellulose fraction, resulting in a reduction in DP (Valjamae et al., 1999).

Amorphous cellulose is prepared by converting the crystalline fraction of cellulose to the amorphous form by mechanical or chemical methods. These celluloses include mechanically made amorphous cellulose, alkaliswollen cellulose, and phosphorous acid swollen cellulose (PASC, Walseth cellulose). Mechanically made amorphous cellulose is often prepared by ball milling or severe blending (Fan et al., 1980; Ghose, 1969; Henrissat et al., 1985; Wood, 1988). Alkaliswollen amorphous cellulose is made by swelling cellulose power in a high concentration of NaOH (e.g., 16% wt/wt) producing the cellulose type II from type I (O'Sullivan, 1997; Wood, 1988). Phosphoric acid swollen cellulose (PASC) is most commonly made by swelling dry cellulose powder by adding 85% ophosphoric acid (Walseth, 1952; Wood, 1988). High concentration phosphoric acid treatment could result in some degree of conversion of type II cellulose from type I (Weimer et al., 1990). The properties of amorphous cellulose made by ball milling, NaOH and H3PO4, vary greatly, depending on cellulose origins, reaction temperature and time, as well as reagent types and concentrations. Therefore, it is nearly impossible to compare hydrolysis rates on various types of amorphous cellulose from different laboratories or even different batches of amorphous cellulose preparations from the same laboratory. Amorphous cellulose should be kept in hydrated condition; simple air-drying dehydration results in a loss of substrate reactivity (Zhang and Lynd, 2004b). The loss of substrate reactivity during dehydration can be minimized through freeze drying or drying after solvent exchange (Fan et al., 1981; Lee et al., 1980). Regenerated cellulose is often made by converting insoluble cellulose to soluble form using cellulose solvents, such as nitric acid, sulfuric acid, ammoniacal cupric hydroxide (Cu(NH3)4(OH)2), N,N-dimethylacetamide (DMAc)/LiCl (Striegel, 1997), and 1-butyl-3methylimidazolium Cl (Swatloski et al., 2002), followed by restoration to physically insoluble form. The major commercial regenerated cellulose is viscose rayon, which is not pure amorphous cellulose due to some recrystallization. Regenerated amorphous cellulose (RAC) can be made by using cold 85% H3PO4 to dissolve cellulose slurry, followed by precipitation with cold water. RAC is a very good homogeneous substrate for cellulase activity assays (Zhang et al., 2006), and is different from Walseth cellulose, prepared from heterogeneous swollen cellulose (Walseth, 1952). RAC has a consistent quality from batch to batch, and is an ideal insoluble nonsubstitutation cellulose substrate for measuring extremely low cellulase activity.

460

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

-Cellulose contains major cellulose and a small amount of hemicellulose. The commercial Sigma cellulose is often used as a reference cellulosic material to evaluate the hydrolysis ability of total cellulase (Kim et al., 2003). Holocellulose is a solid residue of wood (lignocellulose) after removal of lignin; -cellulose is a solid residue of holocellulose after removal of major hemicellulose by alkali extraction (Green, 1963); after the neutralization of soluble alkali extract materials from holocellulose, the insoluble fraction and the soluble fraction are -cellulose and -cellulose, respectively (Corbett, 1963a; Corbett, 1963b). Lignocellulose pretreatment breaks up the recalcitrant structure of lignocellulose so that cellulase can hydrolyze pretreated lignocellulose faster and more efficiently. Current leading lignocellulose pretreatment technologies, including dilute acid, hot water, flow through, ammonia fiber explosion (AFEX), ammonia recycle percolation, and lime, have been recently reviewed elsewhere (Mosier et al., 2005; Wyman et al., 2005a,b). In addition, two other pretreatments steam explosion and organosolvhave been intensively investigated (Arato et al., 2005; Bura et al., 2002, 2003; Galbe and Zacchi, 2002; Ohgren et al., 2005; Pan et al., 2005a,b; Pye and Lora, 1991; Sassner et al., 2005 ; Soderstrom et al., 2003; Wingren et al., 2003). The substrate characteristics (e.g., cellulose accessibility, DP, hemicellulose content, and lignin content) of pretreated lignocelluloses vary greatly, strongly depending on pretreatment methods and severity, and on lignocellulose origins. For example, the goal of AFEX is to break up the linkages among lignin, hemicellulose, and cellulose, but not to remove any main component. Therefore, the addition of hemicellulase into the cellulase mixture would be important for improving overall hydrolysis performance for AFEX-treated feedstock (Teymouri et al., 2005). Dilute acid pretreatment not only to breaks the linkage among lignin, hemicellulose, and cellulose, but also removes major hemicellulose. Therefore, the addition of hemicellulase is not necessary for an improvement in cellulase mixture performance; while the addition of non-hydrolysis proteins (e.g. bovine serum albumin) into the cellulase mixture could reduce the use of cellulase because of minimization of non-hydrolysis adsorption of cellulase to lignin (Pan et al., 2005b; Wyman CE, personal communication). Organosolv pretreatment significantly removes both hemicellulose and lignin (Arato et al., 2005; Pan et al., 2005a; Pye and Lora, 1991). Therefore, neither hemicellulase nor other protein blockers need to be added. A novel cellulose-solvent-based lignocellulose fractionation is

under development by our laboratory; the hydrolysis rates of residual cellulose samples containing little hemicellulose and lignin cannot be improved by the addition of either hemicellulase or non-hydrolysis protein (Zhang et al., unpublished). In a word, improvements in the overall performance of cellulase mixture by cocktailing are strongly dependent on residual lignocellulose properties, and remains in the trial-and-test stage. Dyed cellulose is prepared by mixing cellulose with a variety of dyes, such as Remazol Brilliant Blue (Holtzapple et al., 1984; Wood, 1988), Reactive Orange (Gusakov et al., 1985), Reactive Blue 19 (Yamada et al., 2005), and fluorescent dye 5-(4,6dichlorotriazinyl) aminofluresceinsm (Helbert et al., 2003). Because of large variations in the surface areas of cellulose and the binding conditions, the quantitative relationship between released dye and reducing sugars must be established for each batch of dyed cellulose. Insoluble cellulose derivatives, such as slightly substituted CMC, can be mixed with a variety of dyes, including Cibacron Blue 3GA and Reactive Orange 14 to produce insoluble dyed-CMC (Ten et al., 2004). Insoluble cellulose derivatives can also be chemically substituted with trinitrophenyl groups to produce chromogenic trinitrophenyl-carboxymethyl cellulose (TNP-CMC) and fluorophoric Fluram cellulose (Huang and Tang, 1976). The TNP-CMC has a 25fold greater sensitivity for endoglucanase activity than does the reducing sugar dinitrosalicyclic acid method, and Fluram cellulose gives another 10-fold increase in sensitivity over TNP-CMC (Huang and Tang, 1976). However, an increased substitution of TNP-CMC reduces substrate solubility and impairs cellulase action along -linked chains (Wood and Bhat, 1988). Sometimes, TNP-CMC is a useful substrate for enzyme solutions containing reducing agents when the reducing sugar assay cannot be conducted (Shinmyo et al., 1979). For example, the cellulosome from the anaerobic bacterium C. thermocellum requires the presence of reducing agents such as DTT or cysteine for activity (Johnson et al., 1982a; Morag et al., 1992; Zhang and Lynd, 2003a). 4. Quantitative assays All existing cellulase activity assays can be divided into three types: (1) the accumulation of products after hydrolysis, (2) the reduction in substrate quantity, and (3) the change in the physical properties of substrates.

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

461

4.1. Hydrolysis products The majority of assays involve the accumulation of hydrolysis products, including reducing sugars, total sugars, and chromophores. The most common reducing sugar assays include the dinitrosalicyclic acid (DNS) method (Ghose, 1987; Miller, 1959), the NelsonSomogyi method (Nelson, 1944; Somogyi, 1952), the 2,2-bicinchroninate (BCA) method (Waffenschmidt and Janeicke, 1987; Zhang and Lynd, 2005b), the 4hydroxybenzoylhydrazine (PAHBAH) method (Lever, 1972; Lever et al., 1973), and the ferricyanide methods (Kidby and Davidson, 1973; Park and Johnson, 1949) in Table 3. Total soluble sugars, regardless of their chain lengths, can be measured directly by the phenol-H2SO4 method (Dubois et al., 1956; Zhang and Lynd, 2005b) or the anthrone-H2SO4 method (Roe, 1955; Viles and Silverman, 1949). Glucose can be measured by an enzymatic glucose kit using coupled hexokinase and glucose-6-phosphate dehydrogenase (Zhang and Lynd, 2004a), or HPLC after post-hydrolysis conversion to glucose. Detection ranges of many sugar assays can be modified using two strategies: (1) a further dilution after the color reaction and (2) varying sugar volume per sample prior to the reaction. For example, the DNS method was originally designed for 20600 g reducing sugar per sample (Miller, 1959), but its detection range

can be expanded to samples of 1002500 g, followed by water dilution (Ghose, 1987). The same is true for the Nelson-Somogyi method. The Sigma enzymatic glucose assay kit was designed to measure sugar concentrations from 200 to 5000 g/L using a reaction mixture consisting of a 10-L sample plus a 1000-L enzyme solution. However, its detection limits can be lowered to 4100 g/L using a reaction mixture of 500-L sample plus 500-L 2-fold concentrated enzyme solution (Zhang and Lynd, 2004a). Major reducing sugar assays depend on the reduction of inorganic oxidants such as cupric ions (Cu2+) or ferricyanide, which accepts electrons from the donating aldehyde groups of reducing cellulose chain ends. Their detection ranges vary from less than 1 g per sample to> 2500 g per sample (Table 3). The DNS and NelsonSomogyi methods are two of the most common assays for measuring reducing sugars for cellulase activity assays because of their relatively high sugar detection range (i.e., no sample dilution required) and low interference from cellulase (i.e., no protein removal required). However, the primary drawback for this method is the poor stoichiometric relationship between cellodextrins and the glucose standard (Coward-Kelly et al., 2003; Ghose, 1987; Kongruang et al., 2004; Wood and Bhat, 1988; Zhang and Lynd, 2005b). For example, the results may suffer from an underestimation of cellulase activity when glucose is used as the standard

Table 3 The common colorimetric sugar assays Method Reducing Sugar Assay DNS DNS Nelson-Somogyi Nelson-Somogyi Nelson Ferricyanide-1 Ferricyanide-2 PAHBAH PAHBAH BCA Modified BCA Total Sugar Assay Phenol-H2SO4 Anthrone-H2SO4 Enzymatic Glucose Assay Glucose-HK/PGHD kit Glucose-HK/PGHD kit Sample (mL) Micro Macro Micro Macro Semi-Micro 13 0.5 15 2 2 13 1 0.5 0.01 0.5 1 Reagent (mL) 3 3 2+2 2+2 2 1+5 0.25 1.5 3 0.5 1 G amount (g/sample) 20600 1002500 110 10600 5100 19 0.181.8 0.55 550 0.24.5 0.49 5100 5100 250 250 G concn. (mg/L) 6.7600 2005000 0.210 5300 2.550 0.39 0.181.8 110 5005000 0.49 0.49 10100 10100 2005000 4100 Ref.

Micro Macro

Miller, 1959 Ghose, 1987 Somogyi, 1952 Somogyi, 1952 Nelson, 1944 Park and Johnson, 1949 Kidby and Davidson, 1973 Lever, 1972 Lever, 1972 Waffenschmidt and Janeicke, 1987 Zhang and Lynd, 2005b

1 1

1+5 1+5

Dubois et al., 1956; Zhang and Lynd, 2005b Roe, 1955; Viles and Silverman, 1949

0.01 0.5

1 0.5

Sigma Kit Zhang and Lynd, 2004a

462

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

and -glucosidase is not in excess (Breuil and Saddler, 1985a,b; Schwarz et al., 1988). The ferricyanide, PAHBAH, and BCA methods, having higher sensitivity to reducing sugar, can detect as little as several micrograms per sample, but suffer from non-specific interference from protein. Total carbohydrate assays, including the phenolH2SO4 method and the anthrone-H2SO4 method, offer two obvious advantages as compared with reducing sugar assays: a strict stoichemetic relationship between cellodextrins (glucose equivalent) and the glucose standard, and little or no interference from protein. But they are limited for application to pure celluloses, because any carbohydrates and their derivatives can have strong interference readings. Using an enzymatic glucose assay kit or HPLC can overcome nonspecific readings from other sugars, but this requires an extra stepconversion of longer cellodextrins to glucose. Total loss of substrate can be measured by several means, such as gravimetry and chemical methods. These methods are not as popular as those involving product accumulation because they involve tedious procedures, such as sample centrifugation or filtration followed by drying. Gravimetry should be employed with care, because the standard deviation of this method is strongly associated with sample weight. For example, two samples of 1mg and 100mg weighed by an analytical balance with accuracy of 0.1mg have 10% and 0.1% standard deviation, respectively. Chemical methods for determining substrate loss include the phenol-H2SO4 (Dubois et al., 1956), the anthrone-H2SO4 (Viles and Silverman, 1949), and the K2Cr2O7H2SO4 methods (Wood, 1988) for residual cellulose, and quantitative saccharification for different carbohydrate components (Ruiz and Ehrman, 1996). Measurable physical cellulose properties representing cellulase activity include swollen factor, fiber strength, structure collapse, turbidity, and viscosity. Earlier assays, involving measurement of the physical changes of the residual solid cellulose, are reviewed here for historical interest. Examples of these assays include the swelling factor (measured by alkali uptake) and the reduction in tensile strength of thread and pulp (Oksanen et al., 2000; Wood, 1975). Typically, the lack of sensitivity limits the use of these assays, except on special occasions (Oksanen et al., 2000; Pere et al., 2001; Wong et al., 2000). For example, Toyama et al. measured total cellulase activity based on the time needed to disintegrate a 1 1 cm filter paper square (Wood, 1988). The turbidometric assay measures a reduction in the absorbance of particle suspension during the hydrolysis process (Enari and Niku-Paavola,

1988; Johnson et al., 1982a,b; Nummi et al., 1981), which monitors the overall hydrolysis rate over a long time but does not measure well the initial hydrolysis rate for individual enzymes. Amorphous cellulose is recommended for turbidometric assays (Enari and NikuPaavola, 1988) because crystalline cellulose hydrolysis could lead to an initial absorbance increase (Zhang, unpublished). Viscosimetric determinations have been used as an assay for the initial hydrolysis rate for endoglucanases using soluble cellulose derivatives (Demeester et al., 1976; Hulme, 1988; Manning, 1981; Miller et al., 1960). Application of this method relies on the assumption that the ratio of viscosity-average molecular weight to number-average molecular weight should remain constant during the period of the assay, which may be true only for a short time (Hulme, 1988). This method is also experimentally cumbersome and difficult to automate. 4.2. Cellulase activity assays The two basic approaches to measuring cellulase activity are (1) measuring the individual cellulase (endoglucanases, exoglucanases, and -glucosidases) activities, and (2) measuring the total cellulase activity. In general, hydrolase enzyme activities are expressed in the form of the initial hydrolysis rate for the individual enzyme component within a short time, or the end-point hydrolysis for the total enzyme mixture to achieve a fixed hydrolysis degree within a given time. For cellulase activity assays, there is always a gap between initial cellulase activity assays and final hydrolysis measurement (Sheehan and Himmel, 1999). To be most meaningful, individual cellulase component assays must also be based on a reliable estimation of the amount of individual enzyme component present in the assay. This information permits the calculation of specific activity, i.e., bonds broken per milligram enzyme per unit time. 4.2.1. Endoglucanases Endoglucanases cleave intramolecular -1,4-glucosidic linkages randomly, and their activities are often measured on a soluble high DP cellulose derivative, such as CMC with the lowest ratio of FRN/Fa (Table 1). The modes of actions of endoglucanases and exoglucanases differ in that endoglucanases decrease the specific viscosity of CMC significantly with little hydrolysis due to intramolecular cleavages, whereas exoglucanases hydrolyze long chains from the ends in a processive process (Irwin et al., 1993; Teeri, 1997; Zhang and Lynd, 2004b). Endoglucanase activities can

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

463

be measured based on a reduction in substrate viscosity and/or an increase in reducing ends determined by a reducing sugar assay. Because exoglucanases also increase the number of reducing ends, it is strongly recommended that endoglucanase activities be measured by both methods (viscosity and reducing ends). Because the carboxymethyl substitutions on CMC make some glucosidic bonds less susceptible to enzyme action, a linear relationship between initial hydrolysis rates and serially diluted enzyme solutions requires (1) dilute enzyme preparation, (2) a short incubation period (e.g., 24 min) or a very low enzyme loading, (3) a low DS CMC, and (4) a sensitive reducing sugar assay. Many workers agree that the BCA method for reducing sugar assay is superior to the DNS method (Carcia et al., 1993). For example, the modified BCA method, which is conducted at 75 C to avoid -glucosidic bond cleavage during the assay, delivers a strict stoichiometry for the reducing ends of cellodextrins regardless of sugar chain lengths (Zhang and Lynd, 2005b) and offers a much higher sensitivity as shown in Table 3 (Zhang and Lynd, 2005b). Soluble oligosaccharides and their chromophoresubstituted substrates, such as p-nitrophenyl glucosides and methylumbelliferyl--D-glucosides, are also used to measure endoglucanase activities based on the release of chromophores or the formation of shorter oligosaccharide fragments, which are measured by HPLC or TLC (Bhat et al., 1990; Claeyssens and Aerts, 1992; van Tilbeurgh and Claeyssens, 1985; Zverlov et al., 2002a, 2002b, 2003, 2005). Endoglucanase activities can also be easily detected on agar plates by staining residual polysaccharides (CMC, cellulose) with various dyes because these dyes are adsorbed only by long chains of polysaccharides (Flp and Ponyi, 1997; Hagerman et al., 1985; Jang et al., 2003; Jung et al., 1998; Kim et al., 2000; Murashima et al., 2002a; Piontek et al., 1998; Rescigno et al., 1994; Ten et al., 2004). These methods are semi-quantitative, and are well suited to monitoring large numbers of samples. Precision is limited because of the relationship between the cleared zone diameters and the logarithm of enzyme activities. For example, differences in enzyme activity levels less than 2-fold are difficult to detect by eye (Sharrock, 1988). Unfortunately, most exoglucanase activities are not detected by these methods, since the processive action of exoglucanases is blocked by carboxymethyl substitutions, which prohibits cellulose chain from shortening. The lack of efficient exoglucanase plate screening method explains some of the difficulty in detecting exoglucanase genes cloned from C. thermocellum (Demain et al., 2005).

4.2.2. Exoglucanases Exoglucanases cleave the accessible ends of cellulose molecules to liberate glucose and cellobiose. T. reesei cellobiohydrolase (CBH) I and II act on the reducing and non-reducing cellulose chain ends, respectively (Teeri, 1997; Teeri et al., 1998; Zhang and Lynd, 2004b). Avicel has been used for measuring exoglucanase activity because it has the highest ratio of FNR/Fa among insoluble cellulosic substrates (Table 1). During chromatographic fractionation of cellulase mixtures, enzymes with little activity on soluble CMC, but showing relatively high activity on avicel, are usually identified as exoglucanases. Unfortunately, amorphous cellulose and soluble cellodextrins are substrates for both purified exoglucanases and endoglucanases. Therefore, unlike endoglucanases and glucosidases, there are no substrates specific for exoglucanases within the cellulase mixtures (Sharrock, 1988; Wood and Bhat, 1988). Claeyssens and his coworkers (van Tilbeurgh et al., 1982) found that 4-methylumbelliferyl--D-lactoside was an effective substrate for T. reesei CBH I, yielding lactose and phenol as reaction products, but it was not a substrate for T. reesei CBH II (van Tilbeurgh and Claeyssens, 1985) and some endoglucanases (van Tilbeurgh et al., 1982). T. reesei EG I, structurally homologous to CBH I, also cleaves 4-methylumbelliferyl--D-lactoside, yet these enzymes can be differentiated by adding cellobiose, an inhibitor that strongly suppresses cellobiohydrolase activity (Claeyssens and Aerts, 1992). T. reesei CBH II does not hydrolyze 4methylumbelliferyl--D-aglycones of either glucose or cellobiose units, but does cleave 4-methylumbelliferyl-D-glycosides with longer glucose chains (van Tilbeurgh et al., 1985). Deshpande et al. (1984) reported a selective assay for exoglucanases in the presence of endoglucanases and glucosidases. This assay is based on the following: (1) exoglucanase specifically hydrolyzes the aglyconic bond of p-nitrophenyl--D-cellobioside to yield cellobiose and p-nitrophenol, (2) -glucosidase activity is inhibited by D-glucono-1,5--lactone (Holtzapple et al., 1990), and (3) the influence of exoglucanase hydrolysis activities must be quantified in the assay procedure in the presence of added purified endoglucanases. However, this technique has its own limitations: (1) CBH II activity cannot be measured using p-nitrophenyl--Dcellobioside, (2) the specific activity of the available purified endoglucanases may not be representative of all existing endoglucanases in the mixture, and (3) the product ratio from endoglucanase actions may be influenced by the presence of exoglucanases.

464

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

4.2.3. -D-glucosidases -D-glucosidases hydrolyze soluble cellobiose and other cellodextrins with a DP up to 6 to produce glucose in the aqueous phase. The hydrolysis rates decrease markedly as the substrate DPs increase (Zhang and Lynd, 2004b). The term cellobiase is often misleading due to this key enzyme's broad substrate specificity beyond a DP of 2. -D-glucosidases are very amenable to a wide range of simple sensitive assay methods, based on colored or fluorescent products released from pnitrophenyl -D-1,4-glucopyranoside (Deshpande et al., 1984; Strobel and Russell, 1987), -naphthyl--Dglucopyranoside, 6-bromo-2-naphthyl--D-glucopyranoside (Polacheck et al., 1987), and 4-methylumbelliferyl--D-glucopyranoside (Setlow et al., 2004). Also, -D-glucosidase activities can be measured using cellobiose, which is not hydrolyzed by endoglucanases and exoglucanases, and using longer cellodextrins, which are hydrolyzed by endoglucanases and exoglucanases (Ghose, 1987; Gong et al., 1977; McCarthy et al., 2004; Zhang and Lynd, 2004b). 4.2.4. Total cellulase The total cellulase system consists of endoglucanases, exoglucanases, and -D-glucosidases, all of which hydrolyze crystalline cellulose synergically. Total cellulase activity assays are always measured using insoluble substrates, including pure cellulosic substrates such as Whatman No. 1 filter paper, cotton linter, microcrystalline cellulose, bacterial cellulose, algal cellulose; and cellulose-containing substrates such as dyed cellulose, -cellulose, and pretreated lignocellulose. The heterogeneity of insoluble cellulose and the complexity of the cellulase system cause formidable problems in measuring total cellulase activity. Experimental results show that the heterogeneous structure of cellulose (filter paper and bacterial cellulose) gives rise to a rapid decrease in the hydrolysis rate within a short time (less than an hour), even when the effects of cellulase deactivation and product inhibition are taken into account (Valjamae et al., 1998; Zhang et al., 1999). In an attempt to clarify this situation, a functionally based model has been developed to demonstrate that the degree of synergism between endoglucanase and exoglucanase is influenced by substrate characteristics, experimental conditions, and enzyme loading/composition ratio (Zhang and Lynd, in press). This model clearly suggests the complexity of total cellulase activity assays and infers that it is nearly impossible to apply the results of the total cellulase activity assay measured on one solid substrate to a different solid substrate. This is one of the reasons that the U.S. DOE-sponsored cellulase

development projects, conducted by Genencor International and Novozymes Biotech, tailored cellulase mixture performance based only on an identical sampledilute acid pretreated corn stover substrate that was prepared in the pilot plant of the National Renewable Energy Laboratory (Golden, CO) (Knauf and Moniruzzaman, 2004). The most common total cellulase activity assay is the filter paper assay (FPA) using Whatman No. 1 filter paper as the substrate, which was established and published by the International Union of Pure and Applied Chemistry (IUPAC) (Ghose, 1987). This assay requires a fixed amount (2mg) of glucose released from a 50-mg sample of filter paper (i.e., 3.6% hydrolysis of the substrate), which ensures that both amorphous and crystalline fractions of the substrate are hydrolyzed. A series of enzyme dilution solutions is required to achieve the fixed degree of hydrolysis. The strong points of this assay are (1) it is based on a widely available substrate, (2) it uses a substrate that is moderately susceptible to cellulases, and (3) it is based on a simple procedure (the removal of residual substrate is not necessary prior to the addition of the DNS reagent). However, the FPA is reproduced in most laboratories with some considerable effort and it has long been recognized for its complexity and susceptibility to operators' errors (Coward-Kelly et al., 2003; Decker et al., 2003). Reliability of results could be influenced by (1) the -D-glucosidase level present in the cellulase mixture (Breuil and Saddler, 1985a,b; Schwarz et al., 1988; Sharrock, 1988), because the DNS readings are strongly influenced by the reducing end ratio of glucose, cellobiose, and longer cellodextrins (Ghose, 1987; Kongruang et al., 2004; Wood and Bhat, 1988; Zhang and Lynd, 2005b); (2) the freshness of the DNS reagent, which is often ignored (Miller, 1959); (3) the DNS reaction conditions, such as boiling severity, heat transfer, and reaction time (Coward-Kelly et al., 2003); (4) the variations in substrate weight based on the area size (1 6 cm a strip), because this method does not require substrate excess (i.e., substrate amounts strongly influence enzyme activity) (Griffin, 1973); and (5) filter paper cutting methods, because the different papercutting methods such as paper punching, razoring, or scissoring could lead to different accessible reducing ends of the substrate (Zhang and Lynd, 2005b). Dyed celluloses are widely used for determining sugar inhibition for total cellulase because they avoid the high background interference from added sugars (Gusakov et al., 1985c; Holtzapple et al., 1984; Wood, 1988). Fluorescent-dyed cellulose is also used for the same purpose, and the higher signal per molecule of

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

465

fluorescent dye permits detection of lower cellulase activities. Researchers should consider the following: (1) the calibration curve between dye release and reducing sugar accumulation should be established for each batch of substrate, because dye adsorption depends on cellulosic substrate properties and preparation conditions; (2) the calibration curve works only for a small hydrolysis conversion range, because dye molecules cannot enter into the internal cellulose structure; and (3) the different hydrolysis modes of endoglucanases and exoglucanases have different dye release preferences (Helbert et al., 2003). Using dyed cellulose, Holtzapple et al. (1984) showed that glucose and cellobiose were noncompetitive inhibitors to the T. reesei cellulase. On the contrary, the T. longibrachiatum cellulase was competitively inhibited by cellobiose and glucose (Gusakov et al., 1985c). Some feel that the different inhibition patterns may be attributed to large variations in characteristics of dyed celluloses (Gruno et al., 2004). Cotton fiber, microcrystalline cellulose, bacterial cellulose, and algal cellulose are several other common pure cellulosic substrates. Powder microcrystalline cellulose could become a preferred substrate to replace filter paper because (1) it can be rapidly dispensed volumetrically as a slurry and thus permits robotics methods; (2) it can be easily pelleted by centrifugation, and the total sugars released are measured more exactly by the phenol-H2SO4 method than by the DNS assay; (3) it is a more recalcitrant substrate, yielding a more stringent substrate for total cellulase activity than does filter paper; and (4) activities measured on microcrystalline cellulose could more accurately represent hydrolysis ability on pretreated lignocellulose, because its characteristics are closer to those of pretreated lignocelluloses, based on cellulose accessibility to cellulase and the degree of polymerization (Zhang and Lynd, 2004b). Sigmacell-20, a readily available microcrystalline cellulose powder, could also be a good alternative substrate for a total cellulase activity assay, replacing Whatman No. 1 filter paper. Keep that in mind, some of the pretreated lignocellulose still contains significant amounts of hemicellulose and lignin, while microcrystalline cellulose does not contain hemicellulose and lignin. -Cellulose and pretreated lignocellulose are often used to evaluate the digestibility of commercial cellulase or of a reconstituted cellulase mixture for a prolonged reaction. The primary difference, as compared to cellulase activity assays using model cellulosic substrates, is the time required for assays, which ranges from several minutes to hours for model substrates

(initial hydrolysis rate) to several days for pretreated lignocellulose to obtain the final digestibility (cellulose conversion). Clearly, the presence of hemicellulose and even lignin results in more complexity. Again, the desired outcome of the experiment must indicate the substrate chosen, especially in the case of total cellulase performance. In conclusion, the measurement of isolated individual cellulase activity is relatively easy, but it is still challenging to measure T. reesei CBH I and CBH II activities specifically in the presence of endoglucanases. There is no clear relationship between the hydrolysis rates obtained on soluble substrates and those on insoluble substrates, mainly because of huge differences in substrate accessibility and DP. For insoluble cellulose, it is highly unlikely that any substantial solubilization of crystalline or semicrystalline cellulose will proceed linearly with time, due to varying -glucosidicbond accessibilities and chain end availability for different regions of fibers. Researchers must state clearly all parameters of their assay conditions, and resist temptation to compare their results to those of other researchers using different substrates, assay methods, etc. For example, the specific activity of Thermobifida fusca YX endoglucanase is reported to be at least ten-fold higher than that of T. reesei endoglucanase on soluble CMC (Himmel et al., 1993); however, this activity ratio is not maintained if the assays are performed with insoluble cellulose (Himmel et al., 1999). 5. Cellulase improvement and screening/selection Two strategies are available for improving the properties of individual cellulase components: (1) rational design and (2) directed evolution. 5.1. Rational design Rational design is the earliest approach to protein engineering, was introduced after the development of recombinant DNA methods and site-directed mutagenesis more than 20 years ago, and is still widely used. This strategy requires detailed knowledge of the protein structure, of the structural causes of biological catalysis or structure-based molecular modeling, and of the ideally structurefunction relationship. As shown in Fig. 3, the process of rational design involves (1) choice of a suitable enzyme, (2) identification of the amino acid sites to be changed, based usually on a high resolution crystallographic structure, and (3) characterization of the mutants. The availability of data on the protein

466

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

Protein structure

Structure-based molecular modeling

Site-directed mutagenesis

Repeat
(optional)

Transformation and Expression

Characterization of mutants

Fig. 3. Scheme of rational protein design.

structure of an enzyme or of homologous proteins typically governs the choice of a suitable enzyme for modification. The identification of the region of the protein to be modified generally requires the knowledge of not only the existing function of the region but also the desired modified or new function. The modification of amino acid sequence can be achieved through sitedirected mutagenesis, exchange of elements of secondary structure, and even exchange of whole domains and/ or generation of fusion proteins. The faith in the power of rational design relies on the belief that our current scientific knowledge is sufficient to predict function from structure. But such information of structures and mechanisms is not available for the vast majority of enzymes. Even if the structure and catalysis mechanism of the target enzyme are well characterized, the molecular mutation basis for the desired function may not be achieved (Arnold, 2001). Rational design appears to be a logical method for researchers to examine possible amino acid sites near to the active site or the binding pocket in a 3-dimensional structure (Bornscheuer and Pohl, 2001). But many important enzymatic properties are not localized in a small number of catalytic residues a priori. Indeed, many residues distributed over large parts of the protein often confer important properties. Even when large functional changes can be obtained with a few amino

acid substitutions, it will often be difficult or impossible to discern the specific mutations responsible. For example, a significant increase (10 6 -fold) in the specificity constant (kcat/KM) of aspartate aminotransferase favoring valine requires 17 amino-acid changes, only one of which occurs within the active-site (Benkovic and Mames-Schiffer, 2003). Recently, a successful computational design to convert non-active ribose binding protein to triose phosphate isomerase was based on 1822 mutations and exhibited a 105106 fold activity enhancement (Dwyer et al., 2004). Unfortunately the success of computational models is often limited to well-understood reactions and enzymes. Different from most enzymes catalyzing soluble substrates in the aqueous phase, cellulase acting on insoluble heterogeneous cellulose is a more complex process, involving: (1) the changes in heterogeneous cellulose characteristics during hydrolysis (Banka et al., 1998; Boisset et al., 2000; Chanzy et al., 1983; Din et al., 1991, 1994; Halliwell and Riaz, 1970; Lee et al., 1996, 2000; Saloheimo et al., 2002; Walker et al., 1990, 1992; Wang et al., 2003; Woodward et al., 1992; Zhang and Lynd, 2004b); (2) cellulase diffusion, adsorption, and catalysis on the surface of cellulose, i.e., decreases from a 3-dimension diffusion (in liquid phase) to a 2dimension diffusion (on solid surfaces) (Henis et al., 1988; Katchalski-Katzir et al., 1985) and even 1-

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

467

dimension processivity along cellulose chains for cellobiohydrolases (Teeri, 1997); (3) the non-productive cellulase binding on the cellulose surface (Beldman et al., 1987; Sheehan and Himmel, 1999); and (4) the yet unexplained dynamic interactions among the cellulosebinding module (CBM), the catalytic domain, and a single glucan chain end lifted from the cellulose surface (Skopec et al., 2003). Several excellent reviews summarize numerous studies using site-directed mutagenesis for investigating cellulase mechanisms and improving enzyme properties (Schulein, 2000; Wilson, 2004; Wither, 2001). Not surprisingly, few researchers using site-directed mutagenesis have reported successful examples of significantly higher activity cellulase mutants on insoluble substrates (Escovar-Kousen et al., 2004; Sakon et al., 1996; Zhang et al., 2000a,b; Zhang and Wilson, 1997). One clear example, however, is the report by Baker and coworkers of a 20% improvement in the activity on microcrystalline cellulose of a modified endoglucanase Cel5A from Acidothermus cellulolyticus (Baker et al., 2005). The Cel5A endoglucanase, whose high-resolution crystallographic structure has been available (Sakon et al., 1996), was subjected to a series of mutations designed to alter the chemistry of the product-leaving side of the active site cleft. Using structural information and following a thesis that end product inhibition could be relieved by a substitution of non-aromatic residue at site 245, a mutant (Y245G) was shown to increase KI of cellobiose by 15-fold. However, today there are no general rules for sitedirected mutagenesis strategies for improving cellulase activity on solid cellulase substrates and it remains in a trial-and-test process.

5.2. Directed evolution Our still limited knowledge about the characteristics of insoluble cellulose substrates, the dynamic interactions between cellulases and insoluble substrates, and the complex synergetic and/or competitive relationships among cellulase components, significantly limits rational design for improving cellulase properties, despite increasing understanding of cellulase structures and hydrolysis mechanisms, characterization of cellulose properties, and cellulase adsorption (Bothwell et al., 1997; Bothwell and Walker, 1995; Bourne and Henrissat, 2001; Lynd et al., 2002; Wither, 2001; Zhang and Lynd, 2004b). In 1999, Michael Himmel (Sheehan and Himmel, 1999) wrote: non-informational approaches to protein engineering should be used to complement existing efforts based on informational or rational design strategies in order to ensure success of the DOE cellulase improvement program. One approach to non-informational mutant identification is irrational design using directed evolution. The greatest advantage of directed evolution is that it is independent of knowledge of enzyme structure and of the interactions between enzyme and substrate. The greatest challenge of this method is developing tools to correctly evaluate the performance of mutants generated by recombinant DNA techniques. The success of a directed evolution experiment depends greatly on the method chosen for finding the best mutant enzyme, often stated as you get what you screen for (Hibbert et al., 2005; Schmidt-Dannert, 2001; Schmidt-Dannert and Arnold, 1999) (see Fig. 4). Table 4 lists the published examples of the cellulases with properties altered using directed evolution. Four

Fig. 4. Scheme of directed protein evolution.

468

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

Table 4 List of cellulases and relevant enzymes whose properties have been changed using directed evolution techniques Enzyme Endoglucanase Endoglucanase Endoglucanase Endoglucanase -D-glucosidase -D -glucosidase -D -glucosidase -D -glucosidase -glycosidase Mutated -glucosidase (glycosynthase) Mutated endoglucanase (glycosynthase) Altered property Thermal stability Activity Alkali pH Cold adoption Thermal stability Thermal stability Activity Activity Activity Activity DNA technique Family shuffling DNA shuffling epPCR Family shuffling DNA shuffling epPCR epPCR + Family shuffling epPCR Family shuffling epPCR cassette mutegenesis Screening/Selection Facilitated screening-Congo red + CMC agar Facilitated screening-Congo red + CMC agar Facilitated screening-Congo red + CMC agar Facilitated screening-Congo red + CMC agar Random Screening-chromogenic substrate Random Screening-chromogenic substrate Random Screening-chromogenic substrate Random Screening-coupled to color reaction Random Screening-chromogenic substrate Facilitated Screening-fluorogenic substrate Chemical complementation Ref. Murashima et al., 2002b Kim et al., 2000 Wang et al., 2005 Catcheside et al., 2003 Lebbink et al., 2000 Gonzalez-Blasco et al., 2000 Arrizubieta and Polaina, 2000 McCarthy et al., 2004 Kaper et al., 2002 Kim et al., 2004 Lin et al., 2004

directed evolution examples have been reported for endoglucanases, all of which are identified by facilitated screening on solid plates containing CMC, followed by Congo Red staining (Catcheside et al., 2003; Kim et al., 2000; Murashima et al., 2002a; Wang et al., 2005). Kim et al. (2000) reported that a 5-fold higher specific activity Bacillus subtilis endoglucanase mutant was found by screening cellulase mutants, generated by DNA shuffling and displayed on the surface of E. coli by fusion of the Pseudomonas syringae ice-nucleation protein. Doi et al. (Murashima et al., 2002b) enhanced the thermostability of an endoglucanase by seven-fold using the family shuffling technique based on the parental Clostridium cellulosomal endoglucanasesEngB and EngD. Gao et al. (Wang et al., 2005) found that a T. reesei EG III mutant generated using the errorprone PCR technique and expressed in Saccharyomyces cerevisiae was found to have an optimal pH of 5.4, corresponding to a basic pH shift of 0.6. Another example identified hybrid mutants using the family shuffling technique for T. reesei cel12A and Hypocrea schweinitzii cel12A genes (Catcheside et al., 2003). -D-glucosidase mutants have been reported to be screened blindly using 96-microplate wells because of lack of facilitated screening tools (Arrizubieta and Polaina, 2000; Gonzalez-Blasco et al., 2000; Lebbink et al., 2000; McCarthy et al., 2004). Improvements in the low temperature catalysis (3-fold) for the hyperthermostable Pyrococcus furiosus -D-glucosidase CelB (Lebbink et al., 2000) and the thermostabilities and catalytic efficiencies for the Paenibacillus polymyxa BgblA and BglA were obtained using the chromogenic substrate, p-nitrophenyl--D-glucopyranoside (Arrizu-

bieta and Polaina, 2000; Gonzalez-Blasco et al., 2000). The hydrolysis rate of the Thermotoga neapolitana 1,4-D-glucan -glucohydrolase (GghA) (EC 3.2.1.74) mutant is increased by 31% after error-prone PCR mutagenesis, in which blind screening was based on glucose released from a non-chromogenic substrate (cellobiose) and measured by the coupled reactions of thermostable glucokinase and glucose-6-phosphate dehydrogenase (McCarthy et al., 2004). In another recent example, after DNA family shuffling, a glycosidase mutant was found to display lactose hydrolysis rates 3.5-fold and 8.6-fold higher than the parental P. furiosus CelB and Sulfolobus solfataricus LacS, respectively, where glucose released from lactose was measured using a coupled glucose oxidase and phenol 4-aminophenazone peroxidase reaction (Kaper et al., 2002). In some cases, glycosyl hydrolases, e.g., Agrobacterium sp. -D-glucosidase, can be converted to glycosynthases by site-directed mutagenesis (Mackenzie et al., 1998). There is no intrinsic way to screen or select for glycosynthase activities today. The specific activity of glycosynthase from Agrobacterium sp. -D-glucosidase was improved (Kim et al., 2004) using a novel coupledenzyme assay and screening on solid plates because another endoglucanase releases fluorophores from the fluorogenic product synthesized by glycosynthase (Mayer et al., 2001). Another selection method for a glycosynthase mutant library is the chemical complementation method (Lin et al., 2004), based on the principle that the glycosynthase activity is linked to the transcription of a LEU2 reporter gene, resulting in cell growth dependant on glycosynthase activity. A 5-fold

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

469

higher activity of glycosynthase is obtained using this approach (Lin et al., 2004). Although a number of successful examples using directed evolution for desired cellulases have been published, the largest limitation of all current selection and screening methods is based on soluble substrates. It is still very challenging to design a method to screen or select cellulase mutants using solid cellulosic substrates. 5.3. Screening The screening strategy is a critical step for finding the desired mutants from a large mutant library. Screening can be divided into two categories: (1) facilitated screening, which distinguishes mutants on the basis of distinct phenotypes, such as chromospheres released or halos formed, and (2) random screening, which picks mutants blindly (Taylor et al., 2001). A typical facilitated screening, carried out on solid agar, relies on product solubilization followed by an enzymatic reaction that gives rise to a zone of identity, such as chromophores released from chromogenic substrates. The assays may be coupled to a second enzyme whose product can in turn be easily monitored, as demonstrated by a successful coupling for cytochrome P450 to horseradish peroxidase (Joo et al., 1999a,b). With the help of microscopic plate images, it is feasible to screen a much larger number of clones on solid plates (e.g., several hundreds per cm2) (Delagrave et al., 2001; Joo et al., 1999b; Youvan et al., 1995). Recently, an ultra-high throughput facilitated screening method, based on solid microbeads, has been developed in which single cells containing mutant genes are immobilized on solid beads. After a chromogenic substrate is applied, stronger colored beads containing desired mutants are identified under the microscope (Freeman et al., 2004). Another facilitated screening method, conducted in the liquid phase, applies a flow cytometer for detecting chromospheres released from chromogenic substrates, which are catalyzed by the celldisplayed enzyme. Numerous reviews pertaining to cell surface displayed enzyme library screening by flow cytometers are available elsewhere (Aharoni et al., 2005; Becker et al., 2004; Cohen et al., 2001; Goddard and Reymond, 2004; Lin and Cornish, 2002; Wahler and Reymond, 2001; Wittrup, 2001). Endoglucanase activities are detected easily by examination of halos on solid agar plates using CMC as the substrate, followed by Congo Red staining and washing. Higher hydrolysis rates of mutants usually result in larger halos (in Section 4.2.1). It is not

surprising that all reported endoglucanase examples using directed evolution have been screened using the CMC/Congro Red method (in Section 5.2). It may be operative to screen exoglucanase mutants on solid plates using soluble chromogenic substrates, such as nitrophenol-cellobioside. However, it is worth noting that the best screening methods for endoglucanases and exoglucanases, capable of hydrolyzing insoluble cellulose, must be implemented on insoluble cellulose rather than on soluble cellulose derivatives. Random screening is another choice, if facilitated screening is not available. It is often implemented using 96-well microtiter plates, although some researchers are moving towards 384-well and higher density plate formats with the help of accurate, low-volume dispensing instruments (Sundberg, 2000). For example, Diversa has developed an ultra-throughput screening platform, the Gigamatrix, having 400,000 wells containing only 50 nL of liquid substrate per well (Wolfson, 2005). But the reformatting of 96-well plates into higher density requires high assay sensitivity and high evaporation control. Additional product measurement can be achieved using HPLC, mass spectrometry, capillary electrophoresis, or IR-thermography (Wahler and Reymond, 2001). A number of improved -glycosidase mutants after random mutagenesis are found using 96-well microplates, as reported in Section 5.2. Recently, in order to measure total cellulase activity, the FPA has been miniaturized from a 1.5-ml enzyme solution to 60 L, which is implemented in a 96-microplate well (Xiao et al., 2004). Water evaporation from samples is prevented using a PCR thermocycler having a built-in 105 C hot lid. Also, Decker and coworkers (2003) have developed a high throughput cellulase assay system using 96microplates equipped in a Cyberlabs C400 robotics deck with the substrates such as Whatman No. 1 filter paper disks (0.25in. diameter), Solka-Floc, SigmaCell-20, Avicel PH101 (FMC, Philadelphia, PA), and cotton linters (Fluka/Sigma Aldrich). This custom system has a maximum output of 84 samples per day and produces values that correlate to the traditional FPA. However, no application of these systems to the screening of higher activity cellulases has been reported. Considering the inherent limitations of the FPA (see Section 4.2.4), this automated approach could be benefited by replacing the DNS method with the phenol-H2SO4 method because the latter (1) has a higher sugar sensitivity (Table 3), (2) is independent of oxygen presence (unlike the DNS reagent) (Miller, 1959), especially for miniaturization that has a very high surface/volume ratio, (3) yields a strict stoichiometric relationship between color

470

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

formation and total soluble sugars released, and (4) is thus independent of -D-glucosidase levels. Different from FPA, the recommended method requires centrifugation for soluble sugars and solid cellulose residue prior to the phenol-sulfuric acid assay. 5.4. Selection Selection is always preferred over screening because it has several-order-of-magnitude higher efficiency than screening (Griffithsa et al., 2004; Olsen et al., 2000; Otten and Quax, 2005). However, selection requires a phenotypic functional link between the target gene and its encoding product that confers selective advantage to its producer. This method is often implemented based on the principles of resistance to cytotoxic agents (e.g., antibiotics) (Stemmer, 1994a,b) or of complementation of auxotroph (Griffithsa et al., 2004; Jurgens et al., 2000; Smiley and Benkovic, 1994). Today, selection on solid media in petri dishes (200300 colonies per dish) is commonly used because a large number of mutants can be identified conveniently by visual inspection of growth or zones around the colonies as a consequence of a diffusing product. Recently, more and more attention has been paid to the traditional liquid selection (or enrichment) technique (Sauer et al., 2000). An exceedingly higher cell concentration of 1012 individual cells per liter and a longer cultivation time (generations) allow continuous culture to become a powerful selection system for the ultra-large size of the mutant library even when selective advantages are very small. Experimental selection strategy is associated with (1) the selection ratio, (2) the location of the targeted enzyme, and (3) the solubility of the substrate for the

targeted enzyme. Fig. 5 presents possible selection strategies and real examples. The selection ratio (RS) is defined as l RS mutant lparent in which mutant and parent are the specific growth rates of the beneficial mutant strain and the parental control, respectively, in terms of time (h- 1). RS values could vary from 1 to infinity. A value of one implies no selective advantage for the mutants in growth rates, and a value of infinity implies an essentially infinite selective advantage, e.g., no growth for wild strains in the presence of antibiotic. When RS > 1.2, a selection experiment can be implemented preferentially on solid media because of easy operation. When RS < 1.2, liquid continuous culture could be the only choice, because a limited number of generations (e.g., 25) cannot generate the significant difference detected, but this limitation can be overcome by employing the continuous culture chemostat. The location of the targeted enzyme is another important concern for selection. Selection for intracellular enzymes is relatively easy, because such benefit is confined to the same organism that produces the protein. A typical example is kanamycin selection, in which only bacteria containing the aminoglycoside phosphototransferase (kan) gene expressed intracellularly can grow in the presence of kanamycin. Other examples of carbohydrate-metabolizing enzymes are intracellular -galactosidase (Horiuchi et al., 1962) and cell-membraneassociated lactose permease (Tsen et al., 1996), when lactose is the controlling growth carbon source. A Saccharomyces cerevisiae mutant containing several recombinant key intracellular genes for xylose

Enzyme Location
Intracellular High RS: Yes. Solid Media Extracellular - tethered Extracellular - secreted High RS: Yes. Solid Media e.g. -lactamase gene in the presence of ampicillin Low RS: Yes. Microcolonization e.g. subiltis gene on BSA

Substrate Solubility

Soluble Insoluble

e.g. kan gene in the presence of kanamycin Low RS: Yes. Chemostat e.g. -galactosidase gene on lactose

High RS: No.

High RS: Yes. Solid Media. e.g. Isolation of cellulolytic microorganisms in the presence of cellulose

Low RS: No.

Low RS: Possible. No example

Low RS: No.

Fig. 5. Selection strategies depending on selection ratio (RS), enzyme location, and substrate solubility.

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

471

catabolism has been selected to utilize xylose efficiently under anaerobic conditions after 460 generations (Sonderegger and Sauer, 2003). When a key enzyme is secreted extracellularly, the benefit from this enzyme is shared with others, resulting in cross feeding. A typical example is the growth of plasmid-deficient stains in liquid fermentations when fermentations are prolonged or the formation of micro-colonies on petri plates because the secreted -lactamase degrades ampicillin with time. A novel method, called microcolonization, has been invented to solve cross feeding challenges for a secreted enzyme (Naki et al., 1998). To employ this method, cells are compartmented into hollow fibers and each single colony grows in its own segment. When soluble substrate is fed to each single cell, diffusion of the secreted enzymes and hydrolytic products (cross feeding) is limited spatially, resulting in effective selective pressure. A 5-fold greater protease-producing mutant was obtained using bovine serum albumin as a sole nitrogen source and by this selection technique (Naki et al., 1998). The examples pertaining to the influence of enzyme location on evolution of specific cellulase activity could exist in nature. The genes encoding the secretory T. reesei cellobiohydrolases are thought to have no selection pressure to increase their catalytic efficiency (Divne et al., 1994; Konstantinidis et al., 1993; Sinnott, 1998). The fact that the cell surface associated cellulase from C. thermocellum ( 2.4 IU/mg, measured at substrate-excess conditions) (Zhang and Lynd, 2003a) has several-fold higher specific activity than the secretory T. reesei cellulase ( 0.6 IU/mg, measured at substrate-excess) (Lynd et al., 2002) on crystalline cellulose could be attributed to the enzyme location. (Note, here we do not use the old data that the specific hydrolysis rate of C. thermocellum cellulase on crystalline cellulose (substrate-excess) was 50-fold higher than the T. reesei cellulase hydrolysis (substrate-limited) (Johnson et al., 1982a,b). Another comparative example (cellulase vs. amylase) also suggests the influence of different enzymes' locations on their evolution rates. The different main biological roles (cellulose as a major component of the plant cell wall and starch as an energy reserve) determine that the majority of cellulases and amylases are located extracellularly and intracellularly, respectively. Therefore, it is not surprising that amylases could have experienced higher evolution rates for achieving higher catalytic efficiency. The difference in catalytic efficiencies for two enzymes on soluble substrates is verified by the results from the Sinot laboratory (Sinnott, 1998). Furthermore, if amylases already have

a little higher specific activity than cellulases, less expression of amylase for utilizing substrates could result in a higher evolution rate than cellulases because of different expression levels (Drummond et al., 2005). Therefore, the different enzyme locations and the resulting expression levels could result in different observed catalysis efficiencies for cellulases and amylases over a long evolution. Substrate solubility of the targeted enzyme is also important for selection, but often is ignored. Because insoluble substrates cannot diffuse across cell membranes, selection requires that a microorganism produce extracellular enzymes that hydrolyze insoluble substrates to produce utilized nutrients. On solid plates, an example of this selection is the isolation of cellulolytic microorganisms that produce extracellular cellulases from non-cellulolytic microorganisms using cellulose as the sole carbon source. In liquid medium, Lelieveld (1982) proposed the use of continuous culture for selection of beneficial hydrolase mutants on solid substrates because there is a diffusion layer of hydrolyzed products between solid substrates and the bulk phase, causing the difference in growth rates. Another theoretical model, proposed by Lynd and coworkers (Fan et al., 2005), further predicted that the tethered hydrolase on the cell surface was a prerequisite for selection on an insoluble substrate. If a hydrolase is secreted into the liquid phase, the cross feeding effect leads to little or no selective pressure (Fan et al., 2005). It must be noted that the above selection analysis is based on the positive or neutral gene control assumption that be right for major carbohydrate hydrolases. In nature, cellulase regulation is more complicated, including negative control and global catabolite regulation (Dror et al., 2003a,b, 2005; Han et al., 2004, 2003; Ilmen et al., 1997; Lin and Wilson, 1988; Mach and Zeilinger, 2003; Spiridonov and Wilson, 1998, 2000; Stevenson and Weimer, 2005; Zhang and Lynd, 2005a). Herein we hypothesize that several key requirements for effective cellulase selection experiments are (1) the cellulase gene is expressed extracellularly in a noncellulolysis host, (2) the cellulase is tethered on the cell surface, and (3) the insoluble cellulosic substrate is used as the limiting carbon source. Difficulty may be posed by the inability of such cell-tethered enzymes to properly access the essential cellulose surface, especially in real plant cell wall (microfibril) fractions. However, aggressive particle size reduction and pretreatment may permit adequate selection. Other challenges lie in heterologous expression of cellulases in hosts suitable for selection. For example, considerable efforts have been made to express heterologous

472

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481

cellulase genes in non-hydrolytic microorganisms, including yeasts, Saccharomyces cereisiae (Fujita et al., 2004, 2002; Hong et al., 2003a,b; Ito et al., 2004; Murai et al., 1997, 1998; Penttila et al., 1988; van Rensburg et al., 1994, 1996; 1998), and Pichia pastoris (Boer et al., 2000; Godbole et al., 1999); and bacteria, E. coli (Francisco et al., 1993; Zhou et al., 1999), and Klebsiella oxytoca (Wood and Ingram, 1992; Zhou et al., 2001; Zhou and Ingram, 1999, 2001). Only the noncellulolytic microorganism, K. oxytoca SZ21, can grow on insoluble amorphous cellulose after transformed with heterologous cellulase genes (Zhou and Ingram, 2001). Insufficient cellulase expression or reduced activity of recombinant cellulases are the two largest obstacles to the selection approach for obtaining higher activity cellulase mutants (Boer et al., 2000; Godbole et al., 1999; Lynd et al., 2005). 6. Summary Effective cellulose hydrolysis requires several different kinds of cellulases working synergically. Although numerous soluble substrates have been developed for measuring endoglucanase and -D-glucosidase activities, specific soluble substrates for exoglucanase activity assays lack. It is emphasized that hydrolysis data from soluble substrates cannot yield useful information on hydrolysis of insoluble substrates. The heterogeneity of insoluble celluloses, the complex dynamic interactions between insoluble substrates and cellulase components, and the complexity of cellulase components result in formidable problems in extrapolating the activity measured on one solid substrate to other solid substrates, especially those with biorefinery impact. This point is key to the eventual improvement of cellulases for the conversion of pretreated plant cell walls in energy crops and agricultural residues. Actual screening and selection effort must be based on physically and chemically relevant industrial substrates. This objective is clearly challenging at this time. The past success of rational design campaigns for improving properties of enzymes have been notable, but the results are costly and come far too slowly (Arnold, 2001). Some feel it is still more of an art than a science and is at a heuristic stage (Arnold, 2001; Arnold et al., 2001; Gustafsson et al., 2003). In contrast, directed evolution is sufficiently robust that improved biocatalysts can be obtained independently of knowledge of the protein structure and the interaction between enzymes and substrate. Therefore, we propose that the employment of continuous culture with insoluble cellulosic substrate as the sole carbon source could be a powerful

tool to select higher activity cellulase mutants that are displayed on the cell surface. Acknowledgments This work was made possible in part by support to YHPZ from the Biological Systems Engineering Department of Virginia Polytechnic Institute and State University. Funding from the U.S. Department of Energy Office of the Biomass Program (to MEH), and UT-Battelle (to JRM) also supported this work. References
Aharoni A, Griffiths AD, Tawfik DS. High-throughput screens and selections of enzyme-encoding genes. Curr Opin Chem Biol 2005;9:2106. Angenent LT, Karim K, Al-Dahhan MH, Wrenn BA, DomiguezEspinosa R. Production of bioenergy and biochemicals from industrial and agricultural wastewater. Trends Biotechnol 2004;22: 47785. Arato C, Pye EK, Gjennestad G. The lignol approach to biorefining of woody biomass to produce ethanol and chemicals. Appl Biochem Biotechnol 2005;121/124:87182. Arnold FH. Combinatorial and computational challenges for biocatalyst design. Nature 2001;409:2537. Arnold FH, Wintrode PL, Miyazaki K, Gershenson A. How enzymes adapt: lessons from directed evolution. Trends Biochem Sci 2001;26:1006. Arrizubieta MJ, Polaina J. Increased thermal resistance and modification of the catalytic properties of a beta-glucosidase by random mutagenesis and in vitro recombination. J Biol Chem 2000;275: 288438. Atalla RH, Vanderhart DL. Native cellulose: a composite of two distinct crystalline forms. Science 1984;223:2835. Baker JO, Ehrman CI, Adney WS, Thomas SR, Himmel ME. Hydrolysis of cellulose using ternary mixtures of purified cellulases. Appl Biochem Biotechnol 1998;70/72:395403. Baker JO, McCarley JR, Lovett R, Yu CH, Adney WS, Rignall TR, et al. Catalytically enhanced endocellulase Cel5A from Acidothermus cellulolyticus. Appl Biochem Biotechnol 2005;121124: 12948. Banka RR, Mishra S, Ghose TK. Fibril formation from cellulose by a novel protein from Trichoderma reesei: a non-hydrolytic cellulolytic component? World J Microbiol Biotechnol 1998;14:5518. Barr BK, Holewinski RJ. 4-Methyl-7-thioumbelliferyl-beta-D-cellobioside: a fluorescent, nonhydrolyzable substrate analogue for cellulases. Biochemistry 2002;41:444752. Bayer EA, Morag E, Lamed R. The cellulosomea treasure-trove for biotechnology. Trends Biotechnol 1994;12:37986. Bayer EA, Shimon LJ, Shoham Y, Lamed R. Cellulosomesstructure and ultrastructure. J Struct Biol 1998;124:22134. Bayer EA, Belaich JP, Shoham Y, Lamed R. The cellulosomes: multienzyme machines for degradation of plant cell wall polysaccharides. Annu Rev Microbiol 2004;58:52154. Becker S, Schmoldt HU, Adams TM, Wilhelm S, Kolmar H. Ultrahigh-throughput screening based on cell-surface display and fluorescence-activated cell sorting for the identification of novel biocatalysts. Curr Opin Biotechnol 2004;15:3239.

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Beguin P, Alzari PM. The cellulosome of Clostridium thermocellum. Biochem Soc Trans 1998;26:17885. Beldman G, Voragen AGJ, Rombouts FM, Searle-van Leeuwen MF, Pilnik W. Adsorption and kinetic behavior of purified endoglucanases and exoglucanases from Trichoderma viride. Biotechnol Bioeng 1987;30:2517. Benkovic SJ, Mames-Schiffer S. A perspective on enzyme catalysis. Science 2003;301:1196202. Bera-Maillet C, Arthaud L, Abad P, Rosso M-N. Biochemical characterization of MI-ENG1, a family 5 endoglucanase secreted by the root-knot nematode Meloidogyne incognita. Eur J Biochem 2000;267(11):325563. Berner RA. The long-term carbon cycle, fossil fuels and atmospheric composition. Nature 2003;426:3236. Bhat KM, Hay AJ, Claeyssens M, Wood TM. Study of the mode of action and site-specificity of the endo-(1-4)-beta-D-glucanases of the fungus Penicillium pinophilum with normal, 1-3H-labelled, reduced and chromogenic cello-oligosaccharides. Biochem J 1990;266:3718. Boer H, Teeri TT, Koivula A. Characterization of Trichoderma reesei cellobiohydrolase Cel7A secreted from Pichia pastoris using two different promoters. Biotechnol Bioeng 2000;69:48694. Boisset C, Chanzy H, Henrissat B, Lamed RL, Shoham Y, Bayer EA. Digestion of crystalline cellulose substrates by the Clostridium thermocellum cellulosome: structural and morphological aspects. Biochem J 1999;340:82935. Boisset C, Fraschini C, Schulein M, Henrissat B, Chanzy H. Imaging the enzymatic digestion of bacterial cellulose ribbons reveals the endo character of the cellobiohydrolase Cel6A from Humicola insolens and its mode of synergy with cellobiohydrolase Cel7A. Appl Environ Microbiol 2000;66:144452. Boisset C, Petrequin C, Chanzy H, Henrissat B, Schulein M. Optimized mixtures of recombinant Humicola insolens cellulases for biodegradation of crystalline cellulose. Biotechnol Bioeng 2001;72:33945. Bornscheuer UT, Pohl M. Improved biocatalysts by directed evolution and rational protein design. Curr Opin Chem Biol 2001;5:13743. Bothwell MK, Walker LP. Evalution of parameter estimation methods for estimating cellulase binding constants. Bioresour Technol 1995;53:219. Bothwell MK, Daughhetee SD, Chaua GY, Wilson DB, Walker LP. Binding capacities for thermomonospora fusca E3, E4 and E5, the E3 binding domain, and Trichoderm reesei CBHI on Avicel and Bacterial Microcrystalline cellulose. Bioresour Technol 1997;60: 69178. Bourne Y, Henrissat B. Glycoside hydrolases and glycosyltransferases: families and functional modules. Curr Opin Struct Biol 2001;11: 593600. Breuil C, Saddler JN. Comparison of the 3,5-dinitrosalicylic acid and Nelson-Somogyi methods of assaying for reducing sugars and determining cellulase activity. Enzyme Microb Technol 1985a;7: 32732. Breuil C, Saddler JN. A comparison of various cellulase assay procedures for measuring the cellulolytic activity of Trichoderma harzianum E58. Biochem Soc Trans 1985b;13:44950. Bura R, Mansfield SD, Saddler JN, Bothast RJ. SO2-catalyzed steam explosion of corn fiber for ethanol production. Appl Biochem Biotechnol 2002;98/100:5972. Bura R, Bothast RJ, Mansfield SD, Saddler JN. Optimization of SO2catalyzed steam pretreatment of corn fiber for ethanol production. Appl Biochem Biotechnol 2003;105/108:31935.

473

Caldeira K, Jain AK, Hoffert MI. Climate sensitivity uncertainty and the need for energy without CO2 emission. Science 2003;299: 20524. Carcia E, Johnston D, Whitaker JR, Shoemaker SP. Assessment of endo-1,4-beta-D-glucanase activity by a rapid colorimetric assay using disodium 2,2'-bicinchoninate. J Food Biochem 1993;17: 13545. Catcheside DE, Rasmussen JP, Yeadon PJ, Bowring FJ, Cambareri EB, et al. Diversification of exogenous genes in vivo in Neurospora. Appl Microbiol Biotechnol 2003;62:5449. Chang MM, Chou TYC, Tsao GT. Structure, pretreatment and hydrolysis of cellulose. Adv Biochem Eng 1981;20:1542. Chanzy H, Henrissat B, Vuong R, Schulein M. The action of 1,4-b-Dglucan cellobio-hydrolase on Valonia cellulose microcrystals. An electron microscopic study. FEBS Lett 1983;153:1137. Cherry JR, Fidantsef AL. Directed evolution of industrial enzymes: an update. Curr Opin Biotechnol 2003;14:43843. Chirico WJ, Brown RDJ. Separation of [1-3H]cellooligosaccharides by thin-layer chromatography: assay for cellulolytic enzymes. Anal Biochem 1985;150:26472. Chirico WJ, Brown RDJ. Beta-glucosidase from Trichoderma reesei: substrate-binding region and mode of action on [3H]cellooligosaccharides. Eur J Biochem 1987;165:34351. Claeyssens M, Aerts G. Characterisation of cellulolytic activities in commercial Trichoderma reesei preparations: an approach using small, chromogenic substrates. Bioresour Technol 1992;39: 1436. Cohen N, Abramov S, Dror Y, Freeman A. In vitro enzyme evolution: the screening challenge of isolating the one in a million. Trends Biotechnol 2001;19:50710. Converse AO. Substrate factors limiting enzymatic hydrolysis. In: Saddler JN, editor. Bioconversion of forest and agricultural plant residues. CAB International; 1993. p. 93106. Corbett WM. Determination of the alpha-cellulose content of cotton and wood cellulose. Methods Carbohydr Chem 1963a;3:278. Corbett WM. Purification of cotton cellulose. Methods Carbohydr Chem 1963b;3:34. Coward-Kelly G, Aiello-Mazzari C, Kim S, Granda C, Holtzapple M. Suggested improvements to the standard filter paper assay used to measure cellulase activity. Biotechnol Bioeng 2003;82:7459. Cox PM, Betts RA, Jones CD, Spall SA, Totterdell IJ. Acceleration of global warming due to carbon-cycle feedbacks in a coupled climate model. Nature 2000;408:1847. Das H, Singh SK. Useful byproducts from cellulosic wastes of agriculture and food industry-a critical appraisal. Crit Rev Food Sci Nutr 2004;44:7789. Decker SR, Adney WS, Jennings E, Vinzant TB, Himmel ME. Automated filter paper assay for determination of cellulase activity. Appl Biochem Biotechnol 2003;105-108:689703. Delagrave S, Murphy DJ, Pruss JL, Maffia AMr, Bylina EJ, et al. Application of a very high-throughput digital imaging screen to evolve the enzyme galactose oxidase. Protein Eng 2001;4:2617. Demain AL. Pickles, pectin, and penicillin. Annu Rev Microbiol 2004;58:142. Demain AL, Newcomb M, Wu JHD. Cellulase, clostridia, and ethanol. Microbiol Mol Biol Rev 2005;69:12454. Demeester J, Cooreman W, Bracke M, Lauwers A. Determination of endoglucanase activity by viscosimetry. Biochem Soc Trans 1976;5:11157. Deshpande MV, Eriksson KE, Pettersson LG. An assay for selective determination of exo-1,4,-beta-glucanases in a mixture of cellulolytic enzymes. Anal Biochem 1984;138:4817.

474

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Fan LT, Lee Y-H, Gharpuray MM. The nature of lignocellulosics and their pretreatments for enzymatic hydrolysis. Adv Biochem Eng 1982;23:15787. Fan Z, McBride JE, van Zyl WH, Lynd LR. Theoretical analysis of selection-based strain improvement for microorganisms with growth dependent upon extracytoplasmic enzymes. Biotechnol Bioeng 2005;92:3544. Fierobe HP, Mechaly A, Tardif C, Belaich A, Lamed R, Shoham Y, et al. Design and production of active cellulosome chimeras. Selective incorporation of dockerin-containing enzymes into defined functional complexes. J Biol Chem 2001;276:2125761. Fierobe HP, Bayer EA, Tardif C, Czjzek M, Mechaly A, Belaich A, et al. Degradation of cellulose substrates by cellulosome chimeras. Substrate targeting versus proximity of enzyme components. J Biol Chem 2002;277:4962130. Fierobe HP, Mingardon F, Mechaly A, Belaich A, Rincon M, Pages S, et al. Action of designer cellulosomes on homogeneous versus complex substrates: controlled incorporation of three distinct enzymes into a defined tri-functional scaffoldin. J Biol Chem 2005;280:1632534. Fleming K, Gray DG, Matthews S. Cellulose crystallites. Chemistry 2001;7:18315. Francisco JA, Stathopoulos C, Warren RAJ, Kilburn DG, Georgiou G. Specific adhesion and hydrolysis of cellulose by intact Escherichia coli expressing surface anchored cellulase or cellulose binding domains. Bio/Technology 1993;11:4915. Freeman A, Cohen-Hadar N, Abramov S, Modai-Hod R, Dror Y, Georgiou G. Screening of large protein libraries by the cell immobilized on adsorbed bead approach. Biotechnol Bioeng 2004;86:196200. Fujita Y, Takahashi S, Ueda M, Tanaka A, Okada H, Morikawa Y, et al. Direct and efficient production of ethanol from cellulosic material with a yeast strain displaying cellulolytic enzymes. Appl Environ Microbiol 2002;68:513641. Fujita Y, Ito J, Ueda M, Fukuda H, Kondo A. Synergistic saccharification, and direct fermentation to ethanol, of amorphous cellulose by use of an engineered yeast strain codisplaying three types of cellulolytic enzyme. Appl Environ Microbiol 2004;70: 120712. Flp L, Ponyi T. Rapid screening for endo-b-1,4-glucanase and endob-1,4-mannanase activities and specific measurement using soluble dye-labelled substrates. J Microbiol Methods 1997;29: 1521. Galbe M, Zacchi G. A review of the production of ethanol from softwood. Appl Microbiol Biotechnol 2002;59:61828. Ghose TK. Continuous enzymatic saccharification of cellulose with culture filtrates of Trichoderma viride QM 6a. Biotechnol Bioeng 1969;11:23961. Ghose TK. Measurement of cellulase activities. Pure Appl Chem 1987;59:25768. Godbole S, Decker SR, Nieves RA, Adney WS, Vinzant TB, et al. Cloning and expression of Trichoderma reesei cellobiohydrolase I in Pichia pastoris. Biotechnol Prog 1999;15:82833. Goddard JP, Reymond JL. Enzyme assays for high-throughput screening. Curr Opin Biotechnol 2004;15:31422. Gong C-S, Ladisch MR, Tsao G. Cellobiase from Trichoderma viride: purification, properties, kinetics and mechanism. Biotechnol Bioeng 1977;19:95981. Gonzalez-Blasco G, Sanz-Aparicio J, Gonzalez B, Hermoso JA, Polaina J. Directed evolution of beta-glucosidase A from Paenibacillus polymyxa to thermal resistance. J Biol Chem 2000;275:13012708.

Din N, Gilkes NR, Tekant B, Miller RC, Anthony JR, Warren RAJ, et al. Non-hydrolytic disruption of cellulose fibres by the binding domain of a bacterial cellulase. Bio/Technology 1991;9: 10969. Din N, Damude HG, Gilkes NR, Miller RC, Warren RAJ, Kilburn DG. C1-Cx revisited: intramolecular synergism in a cellulase. Proc Natl Acad Sci U S A 1994;91:113837. Divne C, Stahlberg J, Reinikainen T, Ruohonen L, Pettersson G, Knowles JKC, et al. The three dimensional structure of cellobiohydrolase I from Trichoderma reesei. Science 1994;265: 5248. Doi RH, Kosugi A. Cellulosomes: plant-cell-wall-degrading enzyme complexes. Nat Rev Microbiol 2004;2:54151. Doi RH, Tamaru Y. The Clostridium cellulovorans cellulosome: an enzyme complex with plant cell wall degrading activity. Chem Rec 2001;1:2432. Doi RH, Park JS, Liu CC, Malburg LM, Tamaru Y, Ichiishi A, et al. Cellulosome and noncellulosomal cellulases of Clostridium cellulovorans. Extremophiles 1998;2:5360. Dong XM, Revol J-F, Gray DG. Effect of microcrystallite preparation conditions on the formation of colloid crystals of cellulose. Cellulose 1998;5:1932. Dror TW, Morag E, Rolider A, Bayer EA, Lamed R, Shoham Y. Regulation of the cellulosomal CelS (cel48A) gene of Clostridium thermocellum is growth rate dependent. J Bacteriol 2003a;185: 30428. Dror TW, Rolider A, Bayer EA, Lamed RL, Shoham Y. Regulation of expression of scaffoldin-related genes in Clostridium thermocellum. J Bacteriol 2003b;185:510916. Dror TW, Rolider A, Bayer EA, Lamed R, Shoham Y. Regulation of major cellulosomal endoglucanases of Clostridium thermocellum differs from that of a prominent cellulosomal xylanase. J Bacteriol 2005;187:22616. Drummond DA, Bloom JD, Adami C, Wilke CO, Arnold FH. Why highly expressed proteins evolve slowly. Proc Natl Acad Sci U S A 2005;102:1433843. Dubois B, Gilles KA, Hamilton JK, Rebers PA, Smith F. Colormetric method for determination of sugars and relative substances. Anal Chem 1956;28:3506. Dwyer MA, Looger LL, Hellinga HW. Computational design of a biologically active enzyme. Science 2004;304:196771. Enari TM, Niku-Paavola ML. Nephelometric and turbidometric assay for cellulase. Methods Enzymol 1988;160:11726. Eriksson KE, Pettersson B. A zymogram technique for the detection of carbohydrases. Anal Biochem 1973;56:61820. Escovar-Kousen JM, Wilson DB, Irwin D. Integration of computer modeling and initial studies of site-directed mutagenesis to improve cellulase activity on Cel9A from Thermobifida fusca. Appl Biochem Biotechnol 2004;113-116:28797. Esterbauer H, Steiner W, Labudova I, Hermann A, Hayn M. Production of Trichoderma cellulase in laboratory and pilot scale. Bioresour Technol 1991;36:5165. Falkowski P, Scholes RJ, Boyle E, Canadell J, Canfield D, Elser J, et al. The global carbon cycle: a test of our knowledge of earth as a system. Science 2000;290:2916. Fan LT, Lee Y-H, Beardmore DH. Mechanism of the enzymatic hydrolysis of cellulose: effects of major structural features of cellulose on enzymatic hydrolysis. Biotechnol Bioeng 1980;22: 17799. Fan LT, Lee Y-H, Beardmore DR. The influence of major structural features of cellulose on rate of enzymatic hydrolysis. Biotechnol Bioeng 1981;23:41924.

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Green JW. Wood cellulose. Methods Carbohydr Chem 1963;3:921. Grethlein HE. Pretreatment for enhanced hydrolysis of cellulosic biomass. Biotechnol Adv 1984;2:4362. Griffin HL. Filter paper assay-effect of time and substrate concentration on cellulase activity. Anal Biochem 1973;56:6215. Griffithsa JS, Cheriyanb M, Corbella JB, Pocivavseka L, Fierke CA, Toone EJ. A bacterial selection for the directed evolution of pyruvate aldolases. Bioorg Med Chem 2004;12:406774. Gruno M, Valjamae P, Pettersson G, Johansson G. Inhibition of the Trichoderma reesei cellulases by cellobiose is strongly dependent on the nature of the substrate. Biotechnol Bioeng 2004;86:50311. Gusakov AV, Sinitsyn AP, Gerasimas VB, Savitskene RY, Steponavichus YY. A product inhibition study of cellulases from Trichoderma longibrachiatum using dyed cellulose. J Biotechnol 1985;3:16774. Gustafsson C, Govindarajan S, Minshull J. Putting engineering back into protein engineering: bioinformatic approaches to catalyst design. Curr Opin Biotechnol 2003;14:36670. Hagerman AE, Blau DH, McClure AL. Plate assay for determining the time of production of protease, cellulase and pectinases by germinating fungal spores. Anal Biochem 1985;151:33442. Haight M. Assessing the environmental burdens of anaerobic digestion in comparison to alternative options for managing the biodegradable fraction of municipal solid wastes. Water Sci Technolnolol 2005;52:5539. Hall DO, Rosillo-Calle F, Williams RH, Woods J. Biomass for energy: supply prospects. In: Johansson TB, Kelly H, Reddy AKN, Willian RH, editors. Renewable energy: sources for fuels and electricity. Washington (DC): Island; 1993. Halliwell G, Riaz M. The formation of short fibres from native cellulose by the components of Trichoderma koningii cellulase. Biochem J 1970;116:3542. Hamer G. Solid waste treatment and disposal: effects on public health and environmental safety. Biotechnol Adv 2003;22:719. Han SO, Yukawa H, Inui M, Doi RH. Regulation of expression of cellulosomal cellulase and hemicellulase genes in Clostridium cellulovorans. J Bacteriol 2003;185:606775. Han SO, Cho HY, Yukawa H, Inui M, Doi RH. Regulation of expression of cellulosomes and noncellulosomal (hemi)cellulolytic enzymes in Clostridium cellulovorans during growth on different carbon sources. J Bacteriol 2004;186:421827. Helbert W, Chanzy H, Husum TL, Schulein M, Ernst S. Fluorescent cellulose microfibrils as substrate for the detection of cellulase activity. Biomacromolecules 2003;4:4817. Henis YI, Yaron T, Lamed RL, Rishpon J, Sahar E, Katchalski-Katzir E. Mobility of enzymes on insoluble substrates: the beta-amylasestarch gel system. Biopolymers 1988;27:12338. Henrissat B. Cellulases and their interaction with cellulose. Cellulose 1994;1:16996. Henrissat B, Driguez H, Viet C, Shulein M. Synergism of cellulases from Trichoderma reesei in the degradation of cellulose. Bio/ Technology 1985;3:7226. Hestrin S. Bacterial cellulose. Methods Carbohydr. Chem. Academic Press; 1963. p. 49. Hibbert EG, Baganz F, Hailes HC, Ward JM, Lye GJ, Woodley JM, Dalby PA. Directed evolution of biocatalytic processes. Biomol Eng 2005;22:119. Himmel ME, Adney WS, Baker JO, Nieves RA, Thomas SR. Cellulases: structure, function and applications. In: Wyman CE, editor. Handbook on Bioethanol. 1993. p. 14461. Himmel ME, Ruth MF, Wyman CE. Cellulase for commodity products from cellosic biomass. Curr Opin Biotechnol 1999;10:35864.

475

Hoffert MI, Caldeira K, Jain AK, Haites EF, Harvey LDD, Potter SD, et al. Energy implications of future stabilization of atmospheric CO2 content. Nature 1998;395:8814. Hoffert MI, Caldeira K, Benford G, Criswell DR, Green C, Herzog H, et al. Advanced technology paths to global climate stability: energy for a greenhouse planet. Science 2002;298: 9817. Holt SM, Hartman PA. A zymogram method to detect endoglucanases from Bacillus subtilis, Myrothecium verrucaria and Trichoderma reesei. J Ind Microbiol Biotechnol 1994;13(1):24. Holtzapple MT. Cellulose. In: Macrae R, Robinson RK, Saddler MJ, editors. Encyclopedia of food science food technology and nutrition. London: Academic Press; 1993. p. 75867. Holtzapple MT, Caram H, Humphrey AE. Determining the inhibition constants in the HCH-1 model of cellulose hydrolysis. Biotechnol Bioeng 1984;26:73557. Holtzapple MT, Cognata M, Shu Y, Hendrickson C. Inhibition of Trichoderma reesei cellulase by sugars and solvents. Biotechnol Bioeng 1990;36:27587. Hong J, Tamaki H, Yamamoto K, Kumagai H. Cloning of a gene encoding a thermo-stable endo-beta-1,4-glucanase from Thermoascus aurantiacus and its expression in yeast. Biotechnol Lett 2003a;25:65761. Hong J, Tamaki H, Yamamoto K, Kumagai H. Cloning of a gene encoding thermostable cellobiohydrolase from Thermoascus aurantiacus and its expression in yeast. Appl Microbiol Biotechnol 2003b;63:4250. Horiuchi T, Tomizawa J-I, Novick A. Isolation and properties of bacteria capable of high rates of b-galactosidase synthesis. Biochim Biophys Acta 1962;55:15263. Hsu T-A. Pretreatment of biomass. In: Wyman CE, editor. Handbook on bioethanol. Taylor & Francis; 1996. Huang JS, Tang J. Sensitive assay for cellulase and dextranase. Anal Biochem 1976;75:36977. Huebner A, Ladisch MR, Tsao GT. Preparation of cellodextrin: an engineering approach. Biotechnol Bioeng 1978;20:166977. Hulme MA. Viscosimetric determination of carboxymethylcellulase activity. Methods Enzymol 1988;160:1305. Humphrey AE, Moreira AA, Arminger W, Zabriskie D. Production of single cell protein from cellulose wastes. Biotechnol Bioeng Symp 1977;7:4564. Ilmen M, Saloheimo A, Onnela M-L, Penttila ME. Regulation of cellulase gene expression in the filamentous fungus Trichoderma reesei. Appl Environ Microbiol 1997;63:1298306. Irwin D, Spezio M, Walker LP, Wilson DB. Activity studies of eight purifiied cellulases: specificity, synergism and binding domain effects. Biotechnol Bioeng 1993;42:100213. Ito J, Fujita Y, Ueda M, Fukuda H, Kondo A. Improvement of cellulose-degrading ability of a yeast strain displaying Trichoderma reesei endoglucanase II by recombination of cellulosebinding domains. Biotechnol Prog 2004;20:68891. Jang SJ, Ham MS, Lee JM, Chung SK, Lee HJ, Kim JH, et al. New integration vector using a cellulase gene as a screening marker for Lactobacillus. FEMS Microbiol Lett 2003;224:1915. Jarvis M. Cellulose stacks up. Science 2003;426:6112. Johnson EA, Sakajoh M, Halliwell G, Madia A, Demain AL. Saccharification of complex cellulosic substrates by the cellulase system from Clostridium thermocellum. Appl Environ Microbiol 1982a;43:112532. Johnson EA, Reese ET, Demain AL. Inhibition of Clostridium thermocellum cellulase by end products of cellulolysis. J Appl Biochem 1982b;4:6471.

476

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Kongruang S, Han MJ, Breton CIG, Penner MH. Quantitative analysis of cellulose-reducing ends. Appl Biochem Biotechnol 2004;113116:21331. Konstantinidis AK, Marsden I, Sinnott ML. Hydrolyses of alpha-and beta-cellobiosyl fluorides by cellobiohydrolases of Trichoderma reesei. Biochem J 1993;291:8838. Krassig HA. Cellulose: structure, accessibility, and reactivity. Yverdon, Switzerland: Gordon and Breach Sci. Publishers; 1993. Lebbink JH, Kaper T, Bron P, van der Oost J, de Vos WM. Improving low-temperature catalysis in the hyperthermostable Pyrococcus furiosus beta-glucosidase CelB by directed evolution. Biochemistry 2000;39:365665. Lee Y-H, Fan LT, Fan L-S. Kinetics of hydrolysis of insoluble cellulose by cellulose. Adv Biochem Eng 1980;17:13168. Lee I, Evans BR, Lane LM, Woodward J. Substrate-enzyme interactions in cellulase systems. Bioresour Technol 1996;58: 1639. Lee I, Evans BR, Woodward J. The mechanism of cellulase action on cotton fibers: evidence from atomic force microscopy. Ultramicroscopy 2000;82:21321. Lelieveld HLM. The use of continuous cultures for selection and isolation of microorganisms producing extracellular enzymes adapted to extreme environments. Biotechnol Bioeng 1982;24: 141925. Leschine SB. Cellulose degradation in anaerobic environments. Annu Rev Microbiol 1995;49:399426. Lever M. A new reaction for colorimetric determination of carbohydratesnext term. Anal Biochem 1972;47:2739. Lever M, Powell JC, Killip M, Small CW. A comparison of 4hydroxybenzoic acid hydrazide (PAHBAH) with other reagents for the determination of glucose. J Lab Clin Med 1973;82:64955. Lin H, Cornish VW. Screening and selection methods for large-scale analysis of protein function. Angew Chem Int Ed Engl 2002;41: 440225. Lin E, Wilson DB. Transpription of the celE gene in Thermomonospora fusca. J Bacteriol 1988;170:383842. Lin KW, Ladisch MR, Schaeffer DM, Noller CH, Lechtenberg V, Tsao GT. Review on effect of pretreatment on digestibility of cellulosic materials. AIChE Symp Ser 1981;203:1026. Lin H, Tao H, Cornish VW. Directed evolution of a glycosynthase via chemical complementation. J Am Chem Soc 2004;126: 1505115059. Lynd LR. Overview and evaluation of fuel ethanol from cellulosic biomass: technology, economics, the environment, and policy. Annu Rev Energy Environ 1996;21:40365. Lynd LR, Cushman JH, Nichols RJ, Wyman CE. Fuel ethanol from cellulosic biomass. Science 1991;251:131823. Lynd LR, Wyman CE, Gerngross TU. Biocommodity engineering. Biotechnol Prog 1999;15:77793. Lynd LR, Weimer PJ, van Zyl WH, Pretorius IS. Microbial cellulose utilization: fundamentals and biotechnology. Microbiol Mol Biol Rev 2002;66:50677. Lynd LR, van Zyl WH, McBride JE, Laser M. Consolidated bioprocessing of cellulosic biomass: an update. Curr Opin Biotechnol 2005;16:57783. Lynd LR, Weimer PJ, Wolfaardt G, Zhang Y-HP. Cellulose hydrolysis by Clostridium thermocellum: a microbial perspective. In: Kataeva IA, editor. Cellulosome. Hauppauge (NY) USA: Nova Science Publishers, Inc. in press. [ISBN 1594549508] Mach RL, Zeilinger S. Regulation of gene expression in industrial fungi: Trichoderma. Appl Microbiol Biotechnol 2003;60:51522.

Joo H, Lin Z, Arnold FH. Laboratory evolution of peroxide-mediated cytochrome P450 hydroxylation. Nature 1999a;399:6703. Joo H, Lin Z, Arnold FH. Laboratory evolution of peroxide-mediated cytochrome P450 hydroxylation. Nature 1999b;399:6703. Jung HC, Park JH, Park SH, Lebeault JM, Pan JG. Expression of carboxymethylcellulase on the surface of Escherichia coli using Pseudomonas syringae ice nucleation protein. Enzyme Microb Technol 1998;22:34854. Jurgens C, Strom A, Wegener D, Hettwer S, Wilmanns M, Sterner R. Directed evolution of a (beta alpha)8-barrel enzyme to catalyze related reactions in two different metabolic pathways. Proc Natl Acad Sci U S A 2000;97:992530. Kamm B, Kamm M. Principles of biorefineries. Appl Microbiol Biotechnol 2004;64:13745. Kaper T, Brouns SJ, Geerling AC, De Vos WM, Van der Oost J. DNA family shuffling of hyperthermostable beta-glycosidases. Biochem J 2002;368:46170. Karlsson J, Momcilovic D, Wittgren B, Schulein M, Tjerneld F, Brinkmalm G. Enzymatic degradation of carboxymethyl cellulose hydrolyzed by the endoglucanases Cel5A, Cel7B, and Cel45A from Humicola insolens and Cel7B, Cel12A and Cel45Acore from Trichoderma reesei. Biopolymers 2001;63:3240. Katchalski-Katzir E, Rishpon J, Sahar E, Lamed RL, Henis YI. Enzyme diffusion and action on soluble and insoluble substrate biopolymers. Biopolymers 1985;24:25777. Kidby DK, Davidson DJ. A convenient ferricyanide estimation of reducing sugars in the nanomole range. Anal Biochem 1973;55:3215. Kim E, Irwin DC, Walker LP, Wilson DB. Factorial optimization of a six-cellulase mixture. Biotechnol Bioeng 1998;58:494501. Kim YS, Jung HC, Pan JG. Bacterial cell surface display of an enzyme library for selective screening of improved cellulase variants. Appl Environ Microbiol 2000;66:78893. Kim TH, Kim JS, Sunwoo C, Lee YY. Pretreatment of corn stover by aqueous ammonia. Bioresour Technol 2003;90:3947. Kim YW, Lee SS, Warren RA, Withers SG. Directed evolution of a glycosynthase from Agrobacterium sp. increases its catalytic activity dramatically and expands its substrate repertoire. J Biol Chem 2004;279:4278793. Kirk O, Borchert TV, Fuglsang CC. Industrial enzyme applications. Curr Opin Biotechnol 2002;13:34551. Klein GL, Snodgrass WR. Cellulose. In: Macrae R, Robinson RK, Saddler MJ, editors. Encyclopedia of Food Science, Food Technology and Nutrition. London: Academic Press; 1993. p. 75867. Kleman-Leyer K, Agosin E, Conner AH, Kirk TK. Changes in molecular size distribution of cellulose during attack by white rot and brown rot fungi. Appl Environ Microbiol 1992;58:126670. Kleman-Leyer KM, Gilkes NR, Miller JRC, Kirk TK. Changes in the molecular-size distribution of insoluble celluloses by the action of recombinant Cellulomonas fimi cellulases. Biochem J 1994;302:4639. Kleman-Leyer KM, Siika-Aho M, Teeri TT, Kirk TK. The cellulase endoglucanase I and cellobiohydrolase II of Trichoderma reesei act syngeristically to solubilize native cotton cellulose but not to decrease its molecular size. Appl Environ Microbiol 1996;62:28837. Klemm D, Philipp B, Heinze T, Heinze U, Wagenknecht W. Comprehensive cellulose chemistry: I. Fundementals and analytical methods. Weinheim, Germany: Wiley-VCH; 1998. Knauf M, Moniruzzaman M. Lignocellulosic biomass processing: a perspective. Int Sugar J 2004;106:14750. Knowles J, Lethtovaara P, Reeri TT. Cellulase families and their genes. Trends Biotechnol 1987;5:25561.

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Mackenzie LF, Wang Q, Warren RAJ, Withers SG. Glycosynthases: mutant glycosidases for oligosaccharide synthesis. J Am Chem Soc 1998;120:55834. Manning K. Improved viscometric assay for cellulase. J Biochem Biophys Methods 1981;5:189202. Mansfield SD, Mooney C, Saddler JN. Substrates and enzyme characteristics that limit cellulose hydrolysis. Biotechnol Prog 1999;15:80416. Mayer C, Jakeman DL, Mah M, Karjala G, Gal L, Warren RA, et al. Directed evolution of new glycosynthases from Agrobacterium beta-glucosidase: a general screen to detect enzymes for oligosaccharide synthesis. Chem Biol 2001;8:43743. McCarthy JK, Uzelac A, Davis DF, Eveleigh DE. Improved catalytic efficiency and active site modification of 1,4-beta-D-glucan glucohydrolase A from Thermotoga neapolitana by directed evolution. J Biol Chem 2004;279:11495502. McCleary BV. New chromogenic substrates for the assay of alphaamylase and (1-4)-beta-glucanase. Carbohydr Res 1980;86: 97104. McMillian JD. Pretreatment of lignocellulosic biomass. ACS Ser 1994;566:23492. Melillo JM, Steudler PA, Aber JD, Newkirk K, Lux H, Bowles FP, et al. Soil warming and carbon-cycle feedbacks to the climate system. Science 2002;298:21736. Mielenz JR. Ethanol production from biomass: technology and commercialization status. Curr Opin Microbiol 2001;4:3249. Miller GL. Use of dinitrosalicylic acid reagent for determination of reducing sugar. Anal Chem 1959;31:4268. Miller GL. Micro column chromatographic method for analysis of oligosaccharides. Anal Biochem 1960;2:13340. Miller GL. Cellodextrins. Methods Carbohydr Chem 1963;3:1349. Miller GL, Blum R, Glennon WE, Burton AL. Measurement of carboxymethylcellulase activity. Anal Biochem 1960;1:12732. Millett MA, Baker AJ, Satter LD. Physical and chemical pretreatments for enhancing cellulose saccharification. Biotechnol Bioeng Symp 1976;6:12553. Mingardon F, Perret S, Belaich A, Tardif C, Belaich JP, Fierobe HP. Heterologous production, assembly, and secretion of a minicellulosome by Clostridium acetobutylicum ATCC 824. Appl Environ Microbiol 2005;71:121522. Mohanty AK, Misra M, Hinrichsen G. Biofibres, biodegradable polymers and biocomposites: an overview. Macromol Mater Eng 2000;276-277:124. Morag E, Bayer EA, Lamed RL. Affinity digestion for the near-total recovery of purified cellulosome from Clostridium thermocellum. Enzyme Microb Technol 1992;14:28992. Moreira N. Growing expectations: new technology could turn fuel into a bump crop. Sci News Online 2005;168(14):20924. Mosier N, Wyman CE, Dale BE, Elander RT, Lee YY, Holtzapple M, et al. Features of promising technologies for pretreatment of lignocellulosic biomass. Bioresour Technol 2005;96:67386. Murai T, Ueda M, Atomi H, Shibasaki Y, Kamasawa N, Osumi M, et al. Genetic immobilization of cellulase on the cell surface of Saccharomyces cerevisiae. Appl Microbiol Biotechnol 1997;48: 499503. Murai T, Ueda M, Kawaguchi T, Arai M, Tanaka A. Assimilation of cellooligosaccharides by a cell surface-engineered yeast expressing beta-glucosidase and carboxymethylcellulase from Aspergillus aculeatus. Appl Environ Microbiol 1998;64:485761. Murashima K, Chen CL, Kosugi A, Tamaru Y, Doi RH, Wong SL. Heterologous production of Clostridium cellulovorans engB, using protease-deficient Bacillus subtilis, and preparation of active recombinant cellulosomes. J Bacteriol 2002a;184:7681.

477

Murashima K, Kosugi A, Doi RH. Thermostabilization of cellulosomal endoglucanase EngB from Clostridium cellulovorans by in vitro DNA recombination with non-cellulosomal endoglucanase EngD. Mol Microbiol 2002b;45:61726. Naki D, Paech C, Ganshaw G, Schellenberger V. Selection of a subtilisin-hyperproducing Bacillus in a highly strcutured environment. Appl Microbiol Biotechnol 1998;49:2904. Nelson N. A photometric adaptation of the Somogyi method for the determination of glucose. J Biol Chem 1944;153:37580. Ng TK, Zeikus JG. Synthesis of [14C] cellobiose with Clostridium thermocellum: cellobiose phosphorylase. Appl Environ Microbiol 1986;52:9024. Nishiyama Y, Sugiyama J, Chanzy H, Langan P. Crystal structure and hydrogen bonding system in cellulose Ia from aynchrotron X-ray and neutron fiber diffraction. J Am Chem Soc 2003;125(47): 143006. Notley SM, Pettersson B, Wagberg L. Direct measurement of attractive van der Waals' forces between regenerated cellulose surfaces in an aqueous environment. J Am Chem Soc 2004;126(43):139301. Nummi M, Fox PC, Niku-Passvolva M-L, Enari T-M. Nephelometric and turbidometric assays of cellulase activity. Anal Biochem 1981;116:1336. Ohgren K, Galbe M, Zacchi G. Optimization of steam pretreatment of SO2-impregnated corn stover for fuel ethanol production. Appl Biochem Biotechnol 2005;121/124:105567. Okazaki M, Moo-Young M. Kinetics of enzymatic hydrolysis of cellulose: analytical description of a mechanistic model. Biotechnol Bioeng 1978;20:63763. Oksanen T, Pere J, Paavilainen L, Buchert J, Viikari L. Treatment of recycled kraft pulps with Trichoderma reesei hemicellulases and cellulases. J Biotechnol 2000;78:3948. Olsen M, Iverson B, Georgiou G. High-throughput screening of enzyme libraries. Curr Opin Biotechnol 2000;11:3317. O'Sullivan AC. Cellulose: the structure slowly unravels. Cellulose 1997;4:173207. Otten LG, Quax WJ. Directed evolution: selecting today's biocatalysts. Biomol Eng 2005;22:19. Pan X, Arato C, Gilkes NR, Gregg D, Mabee W, Pye K, et al. Biorefining of softwood using ethanol organosolv pulping: preliminary evaluation of process streams for manufacture of fuel-grade ethanol and co-products. Biotechnol Bioeng 2005a;90:47381. Pan X, Xie D, Gilkes N, Gregg DJ, Saddler JN. Strategies to enhance the enzymatic hydrolysis of pretreated softwood with high residual lignin content. Appl Biochem Biotechnol 2005b;124:106980. Park JT, Johnson MJ. A submicrodetermination of glucose. J Biol Chem 1949;181:14951. Penttila M, Andre L, Lehtovaara P, Bailey M, Teeri TT, Knowles J. Efficient secretion of two fungal cellobiohydrolases by Saccharomyces cerevisiae. Gene 1988;63:10312. Pere J, Puolakka A, Nousiainan P, Buchert J. Action of purified Trichoderma reesei cellulases on cotton fibers and yarn. J Biotechnol 2001;89:24755. Pereira AN, Mobedshahi M, Ladisch MR. Preparation of cellodextrins. Methods Enzymol 1988;160:2643. Piontek M, Hagedorn J, Hollenberg CP, Gellissen G, Strasser AW. Two novel gene expression systems based on the yeasts Schwanniomyces occidentalis and Pichia stipitis. Appl Microbiol Biotechnol 1998;50:3318. Polacheck I, Melamed M, Bercovier H, Salkin IF. beta-Glucosidase in Candida albicans and its application in yeast identification. J Clin Microbiol 1987;25:90710.

478

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Sharrock KR. Cellulase assay methods: a review. J Biochem Biophys Methods 1988;17:81105. Sheehan J, Himmel M. Enzymes, energy, and the environment: a strategic perspective on the U.S. department of energy's research and development activities for bioethanol. Biotechnol Prog 1999;15:81727. Shinmyo A, Garcia-Martinez DV, Demain AL. Studies on the extracellular cellulolytic enzyme complex produced by Clostridium thermocellum. J Appl Biochem 1979;1:2029. Shintate K, Kitaoka M, Kim YK, Hayashi K. Enzymatic synthesis of a library of beta-(1->4) hetero-D-glucose and D-xylose-based oligosaccharides employing cellodextrin phosphorylase. Carbohydr Res 2003;338:198190. Shoemaker HE, Mink D, Wubbolts MG. Dispelling the mythsbiocatalysis in industrial synthesis. Science 2003;299:16947. Sinnott ML. The cellobiohydrolases of Trichoderma reesei: a review of indirect and direct evidence that their function is not just glycosidic bond hydrolysis. Biochem Soc Trans 1998;26:1604. Skopec CE, Himmel ME, Matthews JF, Brady JW. Energetics for displacing a single chain from the surface of microcrystalline cellulose into the active site of Acidothermus cellulolyticus Cel5A. Protein Eng 2003;16:100515. Smiley JA, Benkovic SJ. Selection of catalytic antibodies for a biosynthetic reaction from a combinatorial cDNA library by complementation of an auxotrophic Escherichia coli: antibodies for orotate decarboxylation. Proc Natl Acad Sci U S A 1994;91: 831923. Soderstrom J, Pilcher L, Galbe M, Zacchi G. Combined use of H2SO4 and SO2 impregnation for steam pretreatment of spruce in ethanol production. Appl Biochem Biotechnol 2003;105/108:12740. Somogyi M. Notes on sugar determination. J Biol Chem 1952;195:1923. Sonderegger M, Sauer U. Evolutionary engineering of Saccharomyces cerevisiae. Appl Environ MicrobiolVol nr 69 2003:19908. Spiridonov NA, Wilson DB. Regulation of biosynthesis of individual cellulases in Thermomonospora fusca. J Bacteriol 1998;180: 354952. Spiridonov NA, Wilson DB. A celR mutation affecting transcription of cellulase genes in Thermobifida fusca. J Bacteriol 2000;182: 2525. Srisodsuk M, Kleman-Leyer K, Keranen S, Kirk TK, Teeri TT. Modes of action on cotton and bacterial cellulose of a homologous endoglucanase-exoglucanase pair from Trichoderma reesei. Eur J Chem 1998;251:88592. Stemmer WPC. Rapid evolution of a protein in vitro by DNA shuffling. Nature 1994a;370:38991. Stemmer WPC. DNA shuffling by random fragmentation and reassembly: in vitro recombination for molecular evolution. Proc Natl Acad Sci U S A 1994b;91:1074751. Stevenson DM, Weimer PJ. Expression of 17 genes in Clostridium thermocellum ATCC 27405 during fermentation of cellulose or Cellobiose in continuous culture. Appl Environ Microbiol 2005;71:46728. Striegel AM. Theory and applications of DMAc/LiCl in the analysis of polysacharrides. Carbohydr Polym 1997;34:26774. Strobel HJ, Russell JB. Regulation of beta-glucosidase in Bacteroides ruminicola by a different mechanism: growth rate-dependent derepression. Appl Environ Microbiol 1987;53:250510. Strobel HJ, Caldwell FC, Dawson KA. Carbohydrate transport by the anaerobic thermophilic Clostridium thermocellum LQRI. Appl Environ Microbiol 1995;61:40125.

Pye EK, Lora JH. The Alcell process: a proven alternative to krfat pulping. Tappi J 1991;74(3):1138. Ramos LA, Assaf JM, ElSeoud OA, Frollini E. Influence of the supramolecular structure and physicochemical properties of cellulose on its dissolution in a lithium chloride/N,N-dimethylacetamide solvent system. Biomacromolecules 2005;6:263847. Reddy N, Yang Y. Biofibers from agricultural byproducts for industrial applications. Trends Biotechnol 2005;23:227. Rescigno A, Rinaldi AC, Curreli N, Olianas A, Sanjust E. A dyed substrate for the assay of endo-1,4-beta-glucanases. J Biochem Biophys Methods 1994;28:1239. Roe JH. The determination of sugar in blood and spinal fluid with anthrone reagent. J Biol Chem 1955;212:33543. Ruiz R, Ehrman T. Determination of carbohydrates in biomass by high performance liquid chromatography Laboratory Analytic Procedure LAP-002, 1996. Russell JB, Rychlik JL. Factors that alter rumen microbial ecology. Science 2001;292:111922. Sabathe F, Soucaille P. Characterization of the CipA scaffolding protein and in vivo production of a minicellulosome in Clostridium acetobutylicum. J Bacteriol 2003;185:10926. Saddler JN, Ramos LP, Breuil C. Steam pretreatment of lignocellulosic residues. In: Saddler JN, editor. Bioconversion of forest and agricultural plant residues. CAB International; 1993. p. 7391. Sakon J, Adney W, Himmel M, Thomas S, Karplus P. Crystalline structure of thermostable family 5 endoglucanase EG1 from Acidothermus cellulolyticum in complex with cellotetraose. Biochemistry 1996;35:1064860. Saloheimo M, Paloheimo M, Hakola S, Pere J, Swanson B, Nyyssonen E, et al. Trichoderma reesei protein with sequence similarity to the plant expansins, exhibits disruption activity on cellulosic materials. Eur J Biochem 2002;269:420211. Sassner P, Galbe M, Zacchi G. Steam pretreatment of Salix with and without SO2 impregnation for production of bioethanol. Appl Biochem Biotechnol 2005;121/124:110117. Sauer J, Sigurskjold BW, Christensen U, Frandsen TP, Mirgorodskaya E, Harrison M, et al. Glucoamylase: structure/function relationships, and protein engineering. Biochim Biophys Acta 2000;1543: 27593. Schlamadinger B, Marland G. The role of forest and bioenergy strategies in the global carbon cycle. Biomass Bioenergy 1996;10:275300. Schloss PD, Hay AG, Wilson DB, Gossett JM, Walker LP. Quantifying bacterial population dynamics in compost using 16S rRNA gene probes. Appl Microbiol Biotechnol 2005;66:45763. Schmid G, Bisell M, Wandrey C. Preparation of cellodextrins and isolation of oligomeric side components and their characterization. Anal Biochem 1988;175:57383. Schmidt-Dannert C. Directed evolution of single proteins, metabolic pathways, and viruses. Biochemistry 2001;40:1312536. Schmidt-Dannert C, Arnold FH. Directed evolution of industrial enzymes. Trends Biotechnol 1999;17:1356. Schulein M. Protein engineering of cellulases. Biochim Biophys Acta 2000;1543:23952. Schwarz WH. The cellulosome and cellulose degradation by anaerobic bacteria. Appl Microbiol Biotechnol 2001;56(56):63449. Schwarz W, Chan M, Breuil C, Saddler JN. Comparison of HPLC and colorometric methods for measuring cellulolytic activity. Appl Microbiol Biotechnol 1988;28:398403. Setlow B, Cabrera-Hernandez A, Cabrera-Martinez RM, Setlow P. Identification of aryl-phospho-beta-D-glucosidases in Bacillus subtilis. Arch Microbiol 2004;181:607.

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Sundberg SA. High-throughput and ultra-high-throughput screening: solution-and cell-based approaches. Curr Opin Biotechnol 2000;11:4753. Swatloski RP, Spear SK, Holbrey JD, Rogers RD. Dissolution of cellulose with ionic liquids. J Am Chem Soc 2002;124:49745. Tao H, Cornish VW. Milestones in directed enzyme evolution. Curr Opin Chem Biol 2002;6:85864. Taylor SV, Kast P, Hilvert D. Investigating and engineering enzymes by genetic selection. Angew Chem Int Ed Engl 2001;40:331035. Teeri TT. Crystalline cellulose degradation: new insights into the function of cellobiohydrolases. Trends Biotechnol 1997;15:1607. Teeri TT, Koivula A, Linder M, Wohlfahrt G, Divne C, Jones TA. Trichoderma reesei cellobiohydrolases: why so efficient on crystalline cellulose? Biochem Soc Trans 1998;26:1738. Ten LN, Im W-T, Kim M-K, Kang MS, Lee S-T. Development of a plate technique for screening of polysaccharide-degrading microorganisms by using a mixture of insoluble chromogenic substrates. J Microbiol Methods 2004;56:37582. Teymouri F, Laureano-Perez L, Alizadeh H, Dale BE. Optimization of the ammonia fiber explosion (AFEX) treatment parameters for enzymatic hydrolysis of corn stover. Bioresour Technol 2005;96:20148. Tomme P, Driver DP, Amandoron EA, Miller JRC, Warren RAJ, Kilburn DG. Comparsion of a fungal (family I) and bacterial (family II) cellulose-binding domain. J Bacteriol 1995;177: 435663. Tomme P, Kwan E, Gilkes NR, Kilburn DG, Warren RA. Characterization of CenC, an enzyme from Cellulomonas fimi with both endo-and exoglucanase activities. J Bacteriol 1996;178:421623. Tsen S-D, Lai S-C, Pang C-P, Lee J-I, Wilson TH. Chemostat selection of an Escherichia coli mutant containing permease with enhanced lactose affinity. Biochem Biophys Res Commun 1996;224:3517. Tuohy MG, Walsh DJ, Murray PG, Claeyssens M, Cuffe MM, Savage AV, et al. Kinetic parameters and mode of action of the cellobiohydrolases produced by Talaromyces emersonii. Biochim Biophys Acta 2002;1596:36680. Valjamae P, Sild V, Pettersson G, Johansson G. The initial kinetics of hydrolysis by cellobiohydrolases I and II is consistent with a cellulose surface- erosion model. Eur J Biochem 1998;253: 46975. Valjamae P, Sild V, Pettersson G, Johansson G. Acid hydrolysis of bacterial cellulose reveals different modes of synergistic action between cellobiohydrolase I and endoglucanase I. Eur J Biochem 1999;266:32734. van Beilen JB, Li Z. Enzyme technology: an overview. Curr Opin Biotechnol 2002;13:33842. van Rensburg P, van Zyl WH, Pretorius IS. Expression of the Butyrivibrio fibrisolvens endo-beta-1,4-glucanase gene together with the Erwinia pectate lyase and polygalacturonase genes in Saccharomyces cerevisiae. Curr Genet 1994;27:1722. van Rensburg P, van Zyl WH, Pretorius IS. Co-expression of a Phanerochaete chrysosporium cellobiohydrolase gene and a Butyrivibrio fibrisolvens endo-beta-1,4-glucanase gene in Saccharomyces cerevisiae. Curr Genet 1996;30:24650. van Rensburg P, Van Zyl WH, Pretorius IS. Engineering yeast for efficient cellulose degradation. Yeast 1998;14:6776. van Tilbeurgh H, Claeyssens M. Detection and differentiation of cellulase components using low molecular mass fluorogenic substrates. FEBS Lett 1985;187:2838.

479

van Tilbeurgh H, Claeyssens M, de Bruyne CK. The use of 4methylumbelliferyl and other chromophoric glycosides in the study of cellulolytic enzymes. FEBS Lett 1982;149:1526. van Tilbeurgh H, Pettersson G, Bhikabhai R, De Boeck H, Claeyssens M. Studies of the cellulolytic system of Trichoderma reesei QM 9414. Reaction specificity and thermodynamics of interactions of small substrates and ligands with the 1,4-beta-glucan cellobiohydrolase II. Eur J Biochem 1985;148:32934. van Wyk JP. Biotechnology and the utilization of biowaste as a resource for bioproduct development. Trends Biotechnol 2001;19:1727. Viles FJ, Silverman L. Determination of starch and cellulose with anthrone. Anal Chem 1949;21:9503. Voloch M, Ladisch MR, Cantarlla M, Tsao GT. Preparation of cellodextrins using sulfuric acid. Biotechnol Bioeng 1984;26: 5579. Waffenschmidt S, Janeicke L. Assay of reducing sugars in the nanomole range with 2,2-bicinchoninate. Anal Biochem 1987;165: 33740. Wahler D, Reymond J-L. Novel methods for biocatalyst screening. Curr Opin Chem Biol 2001;5:1528. Walker LP, Wilson DB, Irwin DC. Measuring fragmentation of cellulose by Thermomonospora fusca cellulase. Enzyme Microb Technol 1990;12:37886. Walker LP, Wilson DB, Irwin DC. Fragmentation of cellulose by the major Thermomonospora fusca cellulase cellulases, Trichoderma reesei CBH1 and their mixtures. Biotechnol Bioeng 1992;40: 101926. Walker LP, Belair CD, Wilson DB, Irwin DC. Engineering cellulase mixtures by varying the mole fraction of Thermomonospora fusca E5 and E3, Trichoderma reesei CBH1 and Caldocellum saccharolyticum b-glucosidase. Biotechnol Bioeng 1993;42: 101928. Walseth CS. Occurrence of cellulases in enzyme preparations from microorganisms. TAPPI 1952;35:22833. Wang W, Liu J, Chen G, Zhang Y, Gao P. Function of a low molecular weight peptide from Trichoderma pseudokoningii S38 during cellulose biodegradation. Curr Microbiol 2003;46:3719. Wang T, Liu X, Yu Q, Zhang X, Qu Y, Gao P, et al. Directed evolution for engineering pH profile of endoglucanase III from Trichoderma reesei. Biomol Eng 2005;22:8994. Weil JWP, Kohlmann K, Ladisch MR. Cellulose pretreatments of lignocellulosic substrates. Enzyme Microb Technol 1994;16: 100214. Weimer PJ, Lopez-Guisa JM, French AD. Effect of cellulose fine structure on kinetics of its digestion by mixed ruminal microorganisms in vitro. Appl Environ Microbiol 1990;56: 241921. Wilson DB. Studies of Thermobifida fusca plant cell wall degrading enzymes. Chem Rec 2004;4:7282. Wilson DB, Walker LP. Engineering cellulases. Bioresour Technol 1991;36:979. Wingren A, Galbe M, Zacchi G. Techno-economic evaluation of producing ethanol from softwood: comparison of SSF and SHF and identification of bottlenecks. Biotechnol Prog 2003;19: 110917. Wirth SJ, Wolf GA. Micro-plate colourimetric assaynext term for Endo-acting previous termcellulase, next term xylanase, chitinase, 1,3-b-glucanase and amylase extracted from forest soil horizons. Soil Biol Biochem 1992;24:5119. Wirth TE, Gray CB, Podesta JD. The future of energy policy. Foreign Aff 2003;82:12544.

480

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Clostridium thermocellum batch cultures. Anal Chem 2003a;75: 21927. Zhang Y-HP, Lynd LR. Cellodextrin preparation by mixed-acid hydrolysis and chromatographic separation. Anal Biochem 2003b;322:22532. Zhang Y-HP, Lynd LR. Kinetics and relative importance of phosphorolytic and hydrolytic cleavage of cellodextrins and cellobiose in cell extracts of Clostridium thermocellum. Appl Environ Microbiol 2004a;70:15639. Zhang Y-HP, Lynd LR. Toward an aggregated understanding of enzymatic hydrolysis of cellulose: noncomplexed cellulase systems. Biotechnol Bioeng 2004b;88:797824. Zhang Y-HP, Lynd LR. Regulation of cellulase synthesis in batch and continuous cultures of Clostridium thermocellum. J Bacteriol 2005a;187:99106. Zhang Y-HP, Lynd LR. Determination of the number-average degree of polymerization of cellodextrins and cellulose with application to enzymatic hydrolysis. Biomacromolecules 2005b;6:15105. Zhang Y-HP, Lynd LR. Cellulose utilization by Clostridium thermocellum: bioenergetics and hydrolysis product assimilation. Proc Natl Acad Sci U S A 2005c;102:73215. Zhang Y-HP, Lynd LR. Biosynthesis of radio-labeled cellodextrins by the Clostridium thermocellum cellobiose and cellodextrin phosphorylases for measurement of intracellular sugars. Appl Microbiol Biotechnol 2006;70(1):1239. Zhang Y-HP, Lynd LR. A functionally-based model for hydrolysis of cellulose by fungal cellulase. Biotechnol Bioeng [in press]. doi:10.1002/bit.20906. Zhang S, Wilson DB. Surface residue mutations which change the substrate specificity of Thermomonospora fusca endoglucanase E2. J Biotechnol 1997;57:10113. Zhang S, Wolfgang DE, Wilson DB. Substrate heterogeneity causes the nonlinear kinetics of insoluble cellulose hydrolysis. Biotechnol Bioeng 1999;66:3541. Zhang S, Barr BK, Wilson DB. Effects of noncatalytic residue mutations on substrate specificity and ligand binding of Thermobifida fusca endocellulase cel6A. Eur J Biochem 2000a;267: 24452. Zhang S, Irwin DC, Wilson DB. Site-directed mutation of noncatalytic residues of Thermobifida fusca exocellulase Cel6B. Eur J Biochem 2000b;267:310115. Zhang Y-HP, Cui J-B, Lynd LR, Kuang LR. A transition from cellulose swelling to cellulose dissolution by o-phosphoric acid: evidences from enzymatic hydrolysis and supramolecular structure. Biomacromolecules 2006;7(2):6448. Zhbankov RG. Hydrogen bonds and structure of carbohydrates. J Mol Struct 1992;270:52339. Zhou S, Ingram LO. Engineering endoglucanase-secreting strains of ethanologenic Klebsiella oxytoca P2. J Ind Microbiol Biotechnol 1999;22:6007. Zhou S, Ingram LO. Simultaneous saccharification and fermentation of amorphous cellulose to ethanol by recombinant Klebsiella oxytoca SZ21 without supplemental cellulase. Biotechnol Lett 2001;23:145562. Zhou S, Yomano LP, Saleh AZ, Davis FC, Aldrich HC, Ingram LO. Enhancement of expression and apparent secretion of Erwinia chrysanthemi endoglucanase (encoded by celZ) in Escherichia coli B. Appl Environ Microbiol 1999;65:243945. Zhou S, Davis FC, Ingram LO. Gene integration and expression and extracellular secretion of Erwinia chrysanthemi endoglucanase CelY (celY) and CelZ (celZ) in ethanologenic Klebsiella oxytoca P2. Appl Environ Microbiol 2001;67:614.

Wither SG. Mechanism of glycosyl transferase and hydrolases. Carbohydr Polym 2001;44:32537. Wittrup KD. Protein engineering by cell-surface display. Curr Opin Biotechnol 2001;12:3959. Wolfenden R, Snider MJ. The depth of chemical time and the powder of enzyme as catalysts. Acc Chem Res 2001;34:93845. Wolfgang DE, Wilson DB. Mechanistic studies of active site mutants of Thermomonospora fusca endocellulase E2. Biochemistry 1999;38:974651. Wolfram ML, Dacons JC. The polymer-homogous series of oligosacchardies from cellulose. J Am Chem Soc 1952;74:53313. Wolfson W. Diversa builds a business with designer bacteria. Chem Biol 2005;12:5035. Wong KK, Richardson JD, Mansfield SD. Enzymatic treatment of mechanical pulp fibers for improving papermaking properties. Biotechnol Prog 2000;16:10259. Wood TM. Properties and modes of action of cellulases. Biotechnol Bioeng Symp 1975;5:11137. Wood TM. Preparation of crystalline, amorphous and dyed cellulase substrate. Methods Enzymol 1988;166:1945. Wood TM, Bhat KM. Methods for measuring cellulase activities. Methods Enzymol 1988;160:87117. Wood TM, Garica-Campayo V. Enzymology of cellulose degradation. Biodegradation 1990;1:14761. Wood BE, Ingram LO. Ethanol production from cellobiose, amorphous cellulose, and crystalline cellulose by recombinant Klebsiella oxytoca containing chromosomally integrated Zymomonas mobilis genes for ethanol production and plasmids expressing thermostable cellulase genes from Clostridium thermocellum . Appl Environ Microbiol 1992;58:210310. Woodward J, Affholter KA, Noles KK, Troy NT, Gaslightwala SF. Does cellobiohydrolase II core protein from Trichoderma reesei disperse cellulose macrofibrils? Enzyme Microb Technol 1992;14:62530. Wyman CE. Ethanol from lignocellulosic biomass: technology, economics, and opportunities. Bioresour Technol 1994;50:315. Wyman CE. Biomass ethanol: technical progress, opportunities, and commercial challenges. Annu Rev Energy Environ 1999;24: 189226. Wyman CE. Potential synergies and challenges in refining cellulosic biomass to fuels, chemicals, and power. Biotechnol Prog 2003;19: 254262. Wyman CE, Dale BE, Elander RT, Holtzapple M, Ladisch MR, Lee YY. Coordinated development of leading biomass pretreatment technologies. Bioresour Technol 2005a;96:195966. Wyman CE, Dale BE, Elander RT, Holtzapple M, Ladisch MR, Lee YY. Comparative sugar recovery data from laboratory scale application of leading pretreatment technologies to corn stover. Bioresour Technol 2005b;96:202632. Xiao Z, Storms R, Tsang A. Microplate-based filter paper assay to measure total cellulase activity. Biotechnol Bioeng 2004;88: 8327. Yamada M, Amano Y, Horikawa E, Nozaki K, Kanda T. Mode of action of cellulases on dyed cotton with a reactive dye. Biosci Biotechnol Biochem 2005;69:4550. Youvan DC, Goldman E, Delagrave S, Yang MM. Digital imaging spectroscopy for massively parallel screening of mutants. Methods Enzymol 1995;246:73248. Zhang Y-HP, Lynd LR. Quantification of cell and cellulase mass concentrations during anaerobic cellulose fermentation: development of an ELISA-based method with application to

Y.-H. Percival Zhang et al. / Biotechnology Advances 24 (2006) 452481 Zverlov VV, Fuchs KP, Schwarz WH. Chi18A, the endochitinase in the cellulosome of the thermophilic, cellulolytic bacterium Clostridium thermocellum. Appl Environ Microbiol 2002a;68: 31769. Zverlov VV, Velikodvorskaya GA, Schwarz WH. A newly described cellulosomal cellobiohydrolase, CelO, from Clostridium thermocellum: investigation of the exo-mode of hydrolysis, and binding capacity to crystalline cellulose. Microbiology 2002b;148:24755. Zverlov VV, Velikodvorskaya GA, Schwarz WH. Two new cellulosome components encoded downstream of celI in the genome of

481

Clostridium thermocellum: the non-processive endoglucanase CelN and the possibly structural protein CseP. Microbiology 2003;149:51524. Zverlov VV, Schantz N, Schwarz WH. A major new component in the cellulosome of Clostridium thermocellum is a processive endobeta-1,4-glucanase producing cellotetraose. FEMS Microbiol Lett 2005;249:3538.

You might also like